1
|
Pandey H, Sharma J, Kumar S, Mohan N, Jain V, Dhua AK, Yadav DK, Dubey AK, Choudhury P, Goel P. Chromosomal Microarray Analysis in Spina Bifida: Genetic Heterogeneity and Its Clinical Implications. J Indian Assoc Pediatr Surg 2025; 30:290-295. [PMID: 40406313 PMCID: PMC12094595 DOI: 10.4103/jiaps.jiaps_217_24] [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: 09/20/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 05/26/2025] Open
Abstract
Background The etiology of spina bifida is multifactorial; the phenotype is the end result of both genetic and environmental influences. While whole exome sequencing has identified several pathogenic variants in Indian cohorts, the role of chromosomal imbalances and long contiguous stretches of homozygosity (LCSHs) remains largely unexplored in this population. Chromosomal microarray analysis (CMA) is an important tool that provides insights into such genetic aberrations, making it significant for evaluating patients with spina bifida. Objective To identify LCSHs and chromosomal imbalances in three spina bifida patients through CMA analysis as a pilot investigation. Materials and Methods Genomic DNA was isolated from three spina bifida patients (P1: 10-year-old female, P2: 1-year-old male, and P3: 2.8-year-old male) and subjected to CMA using the Affymetrix 750K high-density array platform. The submicroscopic chromosomal imbalances and LCSHs were cross-referenced with public databases (Database of Genomic Variants, ClinVar, and OMIM) to evaluate their clinical significance. Functional annotations of the affected genes were performed to understand their role in neural tube development. Results CMA revealed significant LCSH on chromosomes 2, 3, and 7 involving the genes SOX11, WNT7A, FZD9, SEMA3A, and VHL, all of which are involved in neural tube closure. Mosaic Klinefelter syndrome (25.9% mosaicism) was identified in the second patient while the third patient had a normal genetic profile. The detection of significant genetic variations in two of three cases underscores the potential utility of CMA in spina bifida patients. Conclusions This study has generated valuable insights into the complex genetic landscape underlying the multifactorial etiopathogenesis of spina bifida. The findings not only underscore the importance of an integrated approach but also support the cause of a platform for large-scale investigations in the Indian population.
Collapse
Affiliation(s)
- Himani Pandey
- Laboratory Head, Redcliffe Laboratories, Noida, Uttar Pradesh, India
| | - Jyoti Sharma
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Sourabh Kumar
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Nakul Mohan
- Laboratory Head, Redcliffe Laboratories, Noida, Uttar Pradesh, India
| | - Vishesh Jain
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Anjan Kumar Dhua
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Devendra Kumar Yadav
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Kumar Dubey
- Department of Biotechnology, Invertis University, Bareilly, Uttar Pradesh, India
| | - Prativa Choudhury
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Prabudh Goel
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
2
|
Saloni, Sachan M, Rahul, Verma RS, Patel GK. SOXs: Master architects of development and versatile emulators of oncogenesis. Biochim Biophys Acta Rev Cancer 2025; 1880:189295. [PMID: 40058508 DOI: 10.1016/j.bbcan.2025.189295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Transcription factors regulate a variety of events and maintain cellular homeostasis. Several transcription factors involved in embryonic development, has been shown to be closely associated with carcinogenesis when deregulated. Sry-like high mobility group box (SOX) proteins are potential transcription factors which are evolutionarily conserved. They regulate downstream genes to determine cell fate, via various signaling pathways and cellular processes essential for tissue and organ development. Dysregulation of SOXs has been reported to promote or suppress tumorigenesis by modulating cellular reprogramming, growth, proliferation, angiogenesis, metastasis, apoptosis, immune modulation, lineage plasticity, maintenance of the stem cell pool, therapy resistance and cancer relapse. This review provides a crucial understanding of the molecular mechanism by which SOXs play multifaceted roles in embryonic development and carcinogenesis. It also highlights their potential in advancing therapeutic strategies aimed at targeting SOXs and their downstream effectors in various malignancies.
Collapse
Affiliation(s)
- Saloni
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Rahul
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Rama Shanker Verma
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| | - Girijesh Kumar Patel
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| |
Collapse
|
3
|
Chen S, Yang J, Qi G, Zhao J, Chen Y, Guo X, Zhang J, Gan J, Jian Z, Jin J. Evodiamine inhibits colorectal cancer cell stemness by disturbing ubiquitin specific protease 4 mediated SOX9 stabilization. Discov Oncol 2025; 16:403. [PMID: 40140224 PMCID: PMC11947356 DOI: 10.1007/s12672-025-02183-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second most deadly cancer worldwide. It is of great importance to explore new mechanisms and therapeutic targets. Ubiquitin-specific protease 4 (USP4), as a key regulator of protein stability, has been proven to be closely associated with cancer progression. In addition, it has been found that evodiamine, a novel alkaloid, can effectively inhibit the stemness of colorectal cancer. Based on this, in this study, we explored in depth the interaction mechanism between USP4 and evodiamine in regulating cancer stemness. Our research data showed that the expression level of USP4 in CRC tissues and cells was significantly increased. Further experiments found that overexpression of USP4 could significantly enhance the migration and invasion of CRC and promote the expression of stemness-related genes such as SOX9, OCT4, and CD133. In contrast, knockdown of USP4 expression in CRC cells had the opposite effect. Moreover, USP4 promotes the progression of CRC by mediating the deubiquitination and stabilization of SOX9 protein. At the same time, evodiamine can significantly inhibit the expression of USP4, SOX9, OCT4, and CD133 in CRC cells. Its mechanism of action lies in the fact that evodiamine disrupts the deubiquitination process of USP4 on SOX9 protein, thereby effectively inhibiting the stemness of CRC. Developing an Evodiamine derivative will provide new approach for improving the outcome of CRC patients.
Collapse
Affiliation(s)
- Siqi Chen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People's Republic of China
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People's Republic of China
- Department of Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Jinfeng Yang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People's Republic of China
- Department of Immunology, Guilin Medical University, Guilin, 541199, People's Republic of China
| | - Guangying Qi
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People's Republic of China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199, People's Republic of China
| | - Jiawei Zhao
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People's Republic of China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199, People's Republic of China
| | - Yujing Chen
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People's Republic of China
- School of Pharmacy, Guilin Medical University, Guilin, 541199, People's Republic of China
| | - Xiaotong Guo
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People's Republic of China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199, People's Republic of China
| | - Juzheng Zhang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People's Republic of China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199, People's Republic of China
| | - Jinfeng Gan
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People's Republic of China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199, People's Republic of China
| | - Zhiyuan Jian
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People's Republic of China.
| | - Jiamin Jin
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People's Republic of China.
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People's Republic of China.
- School of Pharmacy, Guilin Medical University, Guilin, 541199, People's Republic of China.
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199, People's Republic of China.
| |
Collapse
|
4
|
Morabito RD, Tatarakis D, Swick R, Stettnisch S, Schilling TF, Horsfield JA, Martin BL. The ratio of Wnt signaling activity to Sox2 transcription factor levels predicts neuromesodermal fate potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633481. [PMID: 39868081 PMCID: PMC11761523 DOI: 10.1101/2025.01.16.633481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Neuromesodermal progenitors (NMPs) are a vertebrate cell type that contribute descendants to both the spinal cord and the mesoderm. The undifferentiated bipotential NMP state is maintained when both Wnt signaling is active and Sox2 is present. We used transgenic reporter lines to live-image both Wnt activity and Sox2 levels in NMPs and observed a unique cellular ratio in NMPs compared to NMP-derived mesoderm or neural tissue. We used this unique signature to identify the previously unknown anatomical position of a progenitor population that gives rise to the midline tissues of the floor plate of the spinal cord and the mesodermal notochord. Thus, quantification of the active Wnt signaling to Sox2 ratio can be used to predict and identify cells with neuromesodermal potential. We also developed the auxin inducible degron 2 system for use in zebrafish to test the temporal role that Sox2 plays during midline formation. We found ectopic Sox2 in the presence of Wnt activity holds cells in the undifferentiated floor plate/notochord progenitor state, and that degradation of the ectopic Sox2 is required for cells to adopt a notochord fate.
Collapse
Affiliation(s)
- Robert D. Morabito
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| | - David Tatarakis
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Ryan Swick
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| | - Samantha Stettnisch
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Julia A. Horsfield
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- The Maurice Wilkins Centre for Biodiscovery, The University of Auckland, Auckland, New Zealand
- Genetics Otago Research Centre, University of Otago, Dunedin, New Zealand
| | - Benjamin L. Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11733, USA
| |
Collapse
|
5
|
Sun L, Wang F, Wang X, Zhang F, Ma S, Lv J. SATB1 mediated tumor colonization and β-catenin nuclear localization are associated with colorectal cancer progression. Cancer Biol Ther 2024; 25:2320307. [PMID: 38385627 PMCID: PMC10885174 DOI: 10.1080/15384047.2024.2320307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/14/2024] [Indexed: 02/23/2024] Open
Abstract
Colorectal cancer (CRC) is a malignancy with high incidence and poor prognosis. It is urgent to identify valuable biomarkers for early diagnosis and potent therapeutic targets. It has been reported that SATB1 is associated with the malignant progression in CRC. To explore the role of SATB1 in CRC progression and the underlying mechanism, we evaluated the expression of SATB1 in the paired CRC tissues with immunohistochemistry. The results showed that the expression of SATB1 in lymph node metastasis was higher than that in primary lesion, and that in distant organ metastasis was higher than that in primary lesion. The retrospective analysis showed that patients with high expression of SATB1 had a significantly worse prognosis than those with negative and moderate expression. In vitro experiments that employing SATB1 over-expressing and depleted CRC cell lines confirmed that SATB1 contributes to cell proliferation and colonization, while inhibiting cell motility. Furthermore, the tissue immunofluorescence assay, Co-IP and Western blot were conducted to reveal that SATB1 induced translocation of β-catenin and formed a protein complex with it in the nuclei. In conclusion, SATB1 mediated tumor colonization and β-catenin nuclear localization are associated with the malignant progression and poor prognosis of CRC.
Collapse
Affiliation(s)
- Luan Sun
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Feng Wang
- Department of Pathology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| | - Xufei Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Feiying Zhang
- The second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Sujuan Ma
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Jinghuan Lv
- Department of Pathology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
6
|
Wu PV, Fish M, Hazard FK, Zhu C, Vennam S, Walton H, Wagh D, Coller J, Przybyl J, Morri M, Neff N, West RB, Nusse R. A developmental biliary lineage program cooperates with Wnt activation to promote cell proliferation in hepatoblastoma. Nat Commun 2024; 15:10007. [PMID: 39567523 PMCID: PMC11579301 DOI: 10.1038/s41467-024-53802-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
Cancers evolve not only through the acquisition and clonal transmission of somatic mutations but also by epigenetic mechanisms that modify cell phenotype. Here, we use histology-guided and spatial transcriptomics to characterize hepatoblastoma, a childhood liver cancer that exhibits significant histologic and proliferative heterogeneity despite clonal activating mutations in the Wnt/β-catenin pathway. Highly proliferative regions with embryonal histology show high expression of Wnt target genes, the embryonic biliary transcription factor SOX4, and striking focal expression of the growth factor FGF19. In patient-derived tumoroids with constitutive Wnt activation, FGF19 is a required growth signal for FGF19-negative cells. Indeed, some tumoroids contain subsets of cells that endogenously express FGF19, downstream of Wnt/β-catenin and SOX4. Thus, the embryonic biliary lineage program cooperates with stabilized nuclear β-catenin, inducing FGF19 as a paracrine growth signal that promotes tumor cell proliferation, together with active Wnt signaling. In this pediatric cancer presumed to originate from a multipotent hepatobiliary progenitor, lineage-driven heterogeneity results in a functional growth advantage, a non-genetic mechanism whereby developmental lineage programs influence tumor evolution.
Collapse
Affiliation(s)
- Peng V Wu
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| | - Matt Fish
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Florette K Hazard
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Chunfang Zhu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sujay Vennam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hannah Walton
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Population Health, NYC Health + Hospitals, New York, NY, 10004, USA
| | - Dhananjay Wagh
- Stanford Genomics, Stanford University, Stanford, CA, 94305, USA
| | - John Coller
- Stanford Genomics, Stanford University, Stanford, CA, 94305, USA
| | - Joanna Przybyl
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Surgery, McGill University, Montreal, H4A 3J1, QC, Canada
- Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, H4A 3J1, QC, Canada
| | - Maurizio Morri
- Chan Zuckerberg Biohub, Stanford, CA, 94305, USA
- Altos Labs, Redwood City, CA, 94065, USA
| | - Norma Neff
- Chan Zuckerberg Biohub, Stanford, CA, 94305, USA
| | - Robert B West
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Roel Nusse
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
7
|
Majer AD, Hua X, Katona BW. Menin in Cancer. Genes (Basel) 2024; 15:1231. [PMID: 39336822 PMCID: PMC11431421 DOI: 10.3390/genes15091231] [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: 08/11/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
The protein menin is encoded by the MEN1 gene and primarily serves as a nuclear scaffold protein, regulating gene expression through its interaction with and regulation of chromatin modifiers and transcription factors. While the scope of menin's functions continues to expand, one area of growing investigation is the role of menin in cancer. Menin is increasingly recognized for its dual function as either a tumor suppressor or a tumor promoter in a highly tumor-dependent and context-specific manner. While menin serves as a suppressor of neuroendocrine tumor growth, as seen in the cancer risk syndrome multiple endocrine neoplasia type 1 (MEN1) syndrome caused by pathogenic germline variants in MEN1, recent data demonstrate that menin also suppresses cholangiocarcinoma, pancreatic ductal adenocarcinoma, gastric adenocarcinoma, lung adenocarcinoma, and melanoma. On the other hand, menin can also serve as a tumor promoter in leukemia, colorectal cancer, ovarian and endometrial cancers, Ewing sarcoma, and gliomas. Moreover, menin can either suppress or promote tumorigenesis in the breast and prostate depending on hormone receptor status and may also have mixed roles in hepatocellular carcinoma. Here, we review the rapidly expanding literature on the role and function of menin across a broad array of different cancer types, outlining tumor-specific differences in menin's function and mechanism of action, as well as identifying its therapeutic potential and highlighting areas for future investigation.
Collapse
Affiliation(s)
- Ariana D Majer
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xianxin Hua
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
8
|
Bugacov H, Der B, Briantseva BM, Guo Q, Kim S, Lindström NO, McMahon AP. Dose-dependent responses to canonical Wnt transcriptional complexes in the regulation of mammalian nephron progenitors. Development 2024; 151:dev202279. [PMID: 39250420 PMCID: PMC11463962 DOI: 10.1242/dev.202279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/02/2024] [Indexed: 09/11/2024]
Abstract
In vivo and in vitro studies argue that concentration-dependent Wnt signaling regulates mammalian nephron progenitor cell (NPC) programs. Canonical Wnt signaling is regulated through the stabilization of β-catenin, a transcriptional co-activator when complexed with Lef/Tcf DNA-binding partners. Using the GSK3β inhibitor CHIR99021 (CHIR) to block GSK3β-dependent destruction of β-catenin, we examined dose-dependent responses to β-catenin in mouse NPCs, using mRNA transduction to modify gene expression. Low CHIR-dependent proliferation of NPCs was blocked on β-catenin removal, with evidence of NPCs arresting at the G2-M transition. While NPC identity was maintained following β-catenin removal, mRNA-seq identified low CHIR and β-catenin dependent genes. High CHIR activated nephrogenesis. Nephrogenic programming was dependent on Lef/Tcf factors and β-catenin transcriptional activity. Molecular and cellular features of early nephrogenesis were driven in the absence of CHIR by a mutated stabilized form of β-catenin. Chromatin association studies indicate low and high CHIR response genes are likely direct targets of canonical Wnt transcriptional complexes. Together, these studies provide evidence for concentration-dependent Wnt signaling in the regulation of NPCs and provide new insight into Wnt targets initiating mammalian nephrogenesis.
Collapse
Affiliation(s)
- Helena Bugacov
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Balint Der
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
- Department of Urology, Faculty of Medicine, Semmelweis University, Budapest 1082, Hungary
- Institute of Translational Medicine, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Bohdana-Myroslava Briantseva
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Qiuyu Guo
- Discovery Biomarkers, Amgen Research, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Sunghyun Kim
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Nils O. Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
9
|
Zhang J, Zhang W, Liu J, Liu Y, Jiang Y, Ainiwaer A, Chen H, Gu Z, Chen H, Mao S, Guo Y, Xu T, Xu Y, Wu Y, Yao X, Yan Y. SOX7 inhibits the malignant progression of bladder cancer via the DNMT3B/CYGB axis. MOLECULAR BIOMEDICINE 2024; 5:36. [PMID: 39227479 PMCID: PMC11371982 DOI: 10.1186/s43556-024-00198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/22/2024] [Indexed: 09/05/2024] Open
Abstract
Bladder cancer (BCa) stands out as a highly prevalent malignant tumor affecting the urinary system. The Sex determining region Y-box protein family is recognized for its crucial role in BCa progression. However, the effect of Sex determining region Y-box 7 (SOX7) on BCa progression has not been fully elucidated. Herein, RNA-sequencing, western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF) and tissue microarray were utilized to assess SOX7 expression in vitro and in vivo. Additionally, SOX7 expression, prognosis, and SOX7 + cytoglobin (CYGB) score were analyzed using R software. In vitro and vivo experiments were performed with BCa cell lines to validate the effect of SOX7 knockdown and overexpression on the malignant progression of BCa. The results showed that SOX7 exhibits low expression in BCa. It functions in diverse capacities, inhibiting the proliferative, migratory, and invasive capabilities of BCa. In addition, the experimental database demonstrated that SOX7 binds to the promoter of DNA methyltransferase 3 beta (DNMT3B), leading to the transcriptional inhibition of DNMT3B. This subsequently results in a reduced methylation of CYGB promoter, ultimately inhibiting the tumor progression of BCa. SOX7 + CYGB scores were significantly linked to patient prognosis. In conclusion, SOX7 inhibits the malignant progression of BCa via the DNMT3B/CYGB axis. Additionally, the SOX7 + CYGB score is capable of predicting the prognostic outcomes of BCa patients. Therefore, SOX7 and CYGB may play an important role in the progression of bladder cancer, and they can be used as prognostic markers of bladder cancer patients.
Collapse
Affiliation(s)
- Jingcheng Zhang
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Wentao Zhang
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Ji Liu
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Yuchao Liu
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Yufeng Jiang
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
- Department of Urology, Chongming Branch, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Ailiyaer Ainiwaer
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
- Department of Urology, Xinjiang Uygur Autonomous Region, Kashgar Prefecture Second People's Hospital, Kashgar, China
| | - Hanyang Chen
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Zhuoran Gu
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Haotian Chen
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Shiyu Mao
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Yadong Guo
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Tianyuan Xu
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Yunfei Xu
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China.
| | - Yuan Wu
- Department of Urology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China.
| | - Xudong Yao
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China.
| | - Yang Yan
- Department of Urology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
10
|
Wang S, Link F, Munker S, Wang W, Feng R, Liebe R, Li Y, Yao Y, Liu H, Shao C, Ebert MP, Ding H, Dooley S, Weng HL, Wang SS. Retinoic acid generates a beneficial microenvironment for liver progenitor cell activation in acute liver failure. Hepatol Commun 2024; 8:e0483. [PMID: 39023343 PMCID: PMC11262820 DOI: 10.1097/hc9.0000000000000483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/05/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND When massive necrosis occurs in acute liver failure (ALF), rapid expansion of HSCs called liver progenitor cells (LPCs) in a process called ductular reaction is required for survival. The underlying mechanisms governing this process are not entirely known to date. In ALF, high levels of retinoic acid (RA), a molecule known for its pleiotropic roles in embryonic development, are secreted by activated HSCs. We hypothesized that RA plays a key role in ductular reaction during ALF. METHODS RNAseq was performed to identify molecular signaling pathways affected by all-trans retinoid acid (atRA) treatment in HepaRG LPCs. Functional assays were performed in HepaRG cells treated with atRA or cocultured with LX-2 cells and in the liver tissue of patients suffering from ALF. RESULTS Under ALF conditions, activated HSCs secreted RA, inducing RARα nuclear translocation in LPCs. RNAseq data and investigations in HepaRG cells revealed that atRA treatment activated the WNT-β-Catenin pathway, enhanced stemness genes (SOX9, AFP, and others), increased energy storage, and elevated the expression of ATP-binding cassette transporters in a RARα nuclear translocation-dependent manner. Further, atRA treatment-induced pathways were confirmed in a coculture system of HepaRG with LX-2 cells. Patients suffering from ALF who displayed RARα nuclear translocation in the LPCs had significantly better MELD scores than those without. CONCLUSIONS During ALF, RA secreted by activated HSCs promotes LPC activation, a prerequisite for subsequent LPC-mediated liver regeneration.
Collapse
Affiliation(s)
- Sai Wang
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Frederik Link
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Munker
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
- Liver Center Munich, University Hospital, LMU, Munich, Germany
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing You’an Hospital, Capital Medical University, Beijing, China
| | - Rilu Feng
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Roman Liebe
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Magdeburg, Germany
| | - Yujia Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ye Yao
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hui Liu
- Department of Pathology, Beijing You’an Hospital, Affiliated with Capital Medical University, Beijing, China
| | - Chen Shao
- Department of Pathology, Beijing You’an Hospital, Affiliated with Capital Medical University, Beijing, China
| | - Matthias P.A. Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center, Mannheim, Germany
| | - Huiguo Ding
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital, Affiliated with Capital Medical University, Beijing, China
| | - Steven Dooley
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hong-Lei Weng
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Shan-Shan Wang
- Beijing Institute of Hepatology, Beijing You’an Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Dias Da Silva I, Wuidar V, Zielonka M, Pequeux C. Unraveling the Dynamics of Estrogen and Progesterone Signaling in the Endometrium: An Overview. Cells 2024; 13:1236. [PMID: 39120268 PMCID: PMC11312103 DOI: 10.3390/cells13151236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/25/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
The endometrium is crucial for the perpetuation of human species. It is a complex and dynamic tissue lining the inner wall of the uterus, regulated throughout a woman's life based on estrogen and progesterone fluctuations. During each menstrual cycle, this multicellular tissue undergoes cyclical changes, including regeneration, differentiation in order to allow egg implantation and embryo development, or shedding of the functional layer in the absence of pregnancy. The biology of the endometrium relies on paracrine interactions between epithelial and stromal cells involving complex signaling pathways that are modulated by the variations of estrogen and progesterone levels across the menstrual cycle. Understanding the complexity of estrogen and progesterone receptor signaling will help elucidate the mechanisms underlying normal reproductive physiology and provide fundamental knowledge contributing to a better understanding of the consequences of hormonal imbalances on gynecological conditions and tumorigenesis. In this narrative review, we delve into the physiology of the endometrium, encompassing the complex signaling pathways of estrogen and progesterone.
Collapse
Grants
- J.0165.24, 7.6529.23, J.0153.22, 7.4580.21F, 7.6518.21, J.0131.19 Fund for Scientific Research
- FSR-F-2023-FM, FSR-F-2022-FM, FSR-F-2021-FM, FSR-F-M-19/6761 University of Liège
- 2020, 2021, 2022 Fondation Léon Fredericq
Collapse
Affiliation(s)
| | | | | | - Christel Pequeux
- Tumors and Development, Estrogen-Sensitive Tissues and Cancer Team, GIGA-Cancer, Laboratory of Biology, University of Liège, 4000 Liège, Belgium; (I.D.D.S.); (V.W.); (M.Z.)
| |
Collapse
|
12
|
Xue Y, Ruan Y, Wang Y, Xiao P, Xu J. Signaling pathways in liver cancer: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:20. [PMID: 38816668 PMCID: PMC11139849 DOI: 10.1186/s43556-024-00184-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Liver cancer remains one of the most prevalent malignancies worldwide with high incidence and mortality rates. Due to its subtle onset, liver cancer is commonly diagnosed at a late stage when surgical interventions are no longer feasible. This situation highlights the critical role of systemic treatments, including targeted therapies, in bettering patient outcomes. Despite numerous studies on the mechanisms underlying liver cancer, tyrosine kinase inhibitors (TKIs) are the only widely used clinical inhibitors, represented by sorafenib, whose clinical application is greatly limited by the phenomenon of drug resistance. Here we show an in-depth discussion of the signaling pathways frequently implicated in liver cancer pathogenesis and the inhibitors targeting these pathways under investigation or already in use in the management of advanced liver cancer. We elucidate the oncogenic roles of these pathways in liver cancer especially hepatocellular carcinoma (HCC), as well as the current state of research on inhibitors respectively. Given that TKIs represent the sole class of targeted therapeutics for liver cancer employed in clinical practice, we have particularly focused on TKIs and the mechanisms of the commonly encountered phenomena of its resistance during HCC treatment. This necessitates the imperative development of innovative targeted strategies and the urgency of overcoming the existing limitations. This review endeavors to shed light on the utilization of targeted therapy in advanced liver cancer, with a vision to improve the unsatisfactory prognostic outlook for those patients.
Collapse
Affiliation(s)
- Yangtao Xue
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yeling Ruan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yali Wang
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Peng Xiao
- Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Junjie Xu
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China.
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China.
- Zhejiang University Cancer Center, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
13
|
Rao X, Zhang Z, Pu Y, Han G, Gong H, Hu H, Ji Q, Liu N. RSPO3 induced by Helicobacter pylori extracts promotes gastric cancer stem cell properties through the GNG7/β-catenin signaling pathway. Cancer Med 2024; 13:e7092. [PMID: 38581123 PMCID: PMC10997846 DOI: 10.1002/cam4.7092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 02/09/2024] [Accepted: 02/28/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) accounts for the majority of gastric cancer (GC) cases globally. The present study found that H. pylori promoted GC stem cell (CSC)-like properties, therefore, the regulatory mechanism of how H. pylori promotes GC stemness was explored. METHODS Spheroid-formation experiments were performed to explore the self-renewal capacity of GC cells. The expression of R-spondin 3 (RSPO3), Nanog homeobox, organic cation/carnitine transporter-4 (OCT-4), SRY-box transcription factor 2 (SOX-2), CD44, Akt, glycogen synthase kinase-3β (GSK-3β), p-Akt, p-GSK-3β, β-catenin, and G protein subunit gamma 7 (GNG7) were detected by RT-qPCR, western blotting, immunohistochemistry (IHC), and immunofluorescence. Co-immunoprecipitation (CoIP) and liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) were performed to identify proteins interacting with RSPO3. Lentivirus-based RNA interference constructed short hairpin (sh)-RSPO3 GC cells. Small interfering RNA transfection was performed to inhibit GNG7. The in vivo mechanism was verified using a tumor peritoneal seeding model in nude mice. RESULTS H. pylori extracts promoted a CSC-like phenotype in GC cells and elevated the expression of RSPO3. RSPO3 knockdown significantly reduced the CSC-like properties induced by H. pylori. Previous studies have demonstrated that RSPO3 potentiates the Wnt/β-catenin signaling pathway, but the inhibitor of Wnt cannot diminish the RSPO3-induced activation of β-catenin. CoIP and LC-MS/MS revealed that GNG7 is one of the transmembrane proteins interacting with RSPO3, and it was confirmed that RSPO3 directly interacted with GNG7. Recombinant RSPO3 protein increased the phosphorylation level of Akt and GSK-3β, and the expression of β-catenin in GC cells, but this regulatory effect of RSPO3 could be blocked by GNG7 knockdown. Of note, GNG7 suppression could diminish the promoting effect of RSPO3 to CSC-like properties. In addition, RSPO3 suppression inhibited MKN45 tumor peritoneal seeding in vivo. IHC staining also showed that RSPO3, CD44, OCT-4, and SOX-2 were elevated in H. pylori GC tissues. CONCLUSION RSPO3 enhanced the stemness of H. pylori extracts-infected GC cells through the GNG7/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xiwu Rao
- Department of OncologyThe First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Postdoctoral Research Station of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Zhipeng Zhang
- Department of OncologyThe First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Postdoctoral Research Station of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Yunzhou Pu
- Department of OncologyShuguang Hospital, Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Gang Han
- Department of OncologyShuguang Hospital, Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hangjun Gong
- Department of GastroenterologyShuguang Hospital, Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hao Hu
- Department of GastroenterologyShuguang Hospital, Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Qing Ji
- Department of OncologyShuguang Hospital, Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Ningning Liu
- Department of OncologyShuguang Hospital, Shanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
14
|
Ling RE, Cross JW, Roy A. Aberrant stem cell and developmental programs in pediatric leukemia. Front Cell Dev Biol 2024; 12:1372899. [PMID: 38601080 PMCID: PMC11004259 DOI: 10.3389/fcell.2024.1372899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/11/2024] [Indexed: 04/12/2024] Open
Abstract
Hematopoiesis is a finely orchestrated process, whereby hematopoietic stem cells give rise to all mature blood cells. Crucially, they maintain the ability to self-renew and/or differentiate to replenish downstream progeny. This process starts at an embryonic stage and continues throughout the human lifespan. Blood cancers such as leukemia occur when normal hematopoiesis is disrupted, leading to uncontrolled proliferation and a block in differentiation of progenitors of a particular lineage (myeloid or lymphoid). Although normal stem cell programs are crucial for tissue homeostasis, these can be co-opted in many cancers, including leukemia. Myeloid or lymphoid leukemias often display stem cell-like properties that not only allow proliferation and survival of leukemic blasts but also enable them to escape treatments currently employed to treat patients. In addition, some leukemias, especially in children, have a fetal stem cell profile, which may reflect the developmental origins of the disease. Aberrant fetal stem cell programs necessary for leukemia maintenance are particularly attractive therapeutic targets. Understanding how hijacked stem cell programs lead to aberrant gene expression in place and time, and drive the biology of leukemia, will help us develop the best treatment strategies for patients.
Collapse
Affiliation(s)
- Rebecca E. Ling
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Joe W. Cross
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Anindita Roy
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Haematology, Great Ormond Street Hospital for Children, London, United Kingdom
| |
Collapse
|
15
|
Wu D, Bai D, Yang M, Wu B, Xu W. Role of Sox9 in BPD and its effects on the Wnt/β-catenin pathway and AEC-II differentiation. Cell Death Discov 2024; 10:20. [PMID: 38212314 PMCID: PMC10784471 DOI: 10.1038/s41420-023-01795-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/24/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024] Open
Abstract
The excessive activation of the Wnt/β-catenin signaling pathway is an important regulatory mechanism that underlies the excessive proliferation and impaired differentiation of type 2 alveolar epithelial cells (AEC-II) in bronchopulmonary dysplasia (BPD). Sox9 has been shown to be an important repressor of the Wnt/β-catenin signaling pathway and plays an important regulatory role in various pathophysiological processes. We found that the increased expression of Sox9 in the early stages of BPD could downregulate the expression of β-catenin and promote the differentiation of AEC-II cells into AEC-I, thereby alleviating the pathological changes in BPD. The expression of Sox9 in BPD is regulated by long noncoding RNA growth arrest-specific 5. These findings may provide new targets for the early intervention of BPD.
Collapse
Affiliation(s)
- Di Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Intensive Care unit, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongqin Bai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Miao Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bo Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
16
|
Shiraishi K, Morley MP, Jones DL, Zhao G, Weiner AI, Basil MC, Cantu E, Ferguson LT, Oyster M, Babu A, Ying Y, Zhou S, Li S, Vaughan AE, Morrisey EE. Airway epithelial cell identity and plasticity are constrained by Sox2 during lung homeostasis, tissue regeneration, and in human disease. NPJ Regen Med 2024; 9:2. [PMID: 38182591 PMCID: PMC10770358 DOI: 10.1038/s41536-023-00344-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 12/14/2023] [Indexed: 01/07/2024] Open
Abstract
Maintenance of the cellular boundary between airway and alveolar compartments during homeostasis and after injury is essential to prohibit pathological plasticity which can reduce respiratory function. Lung injury and disease can induce either functional alveolar epithelial regeneration or dysplastic formation of keratinized epithelium which does not efficiently contribute to gas exchange. Here we show that Sox2 preserves airway cell identity and prevents fate changes into either functional alveolar tissue or pathological keratinization following lung injury. Loss of Sox2 in airway epithelium leads to a loss of airway epithelial identity with a commensurate gain in alveolar and basal cell identity, in part due to activation of Wnt signaling in secretory cells and increased Trp63 expression in intrapulmonary basal-like progenitors. In idiopathic pulmonary fibrosis, loss of SOX2 expression correlates with increased WNT signaling activity in dysplastic keratinized epithelium. SOX2-deficient dysplastic epithelial cells are also observed in COVID-19 damaged lungs. Thus, Sox2 provides a molecular barrier that suppresses airway epithelial plasticity to prevent acquisition of alveolar or basal cell identity after injury and help guide proper epithelial fate and regeneration.
Collapse
Affiliation(s)
- Kazushige Shiraishi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael P Morley
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dakota L Jones
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gan Zhao
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Aaron I Weiner
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Maria C Basil
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Edward Cantu
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Laura T Ferguson
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michele Oyster
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Apoorva Babu
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yun Ying
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Su Zhou
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shanru Li
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Andrew E Vaughan
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
17
|
Guo R, Guo YZ, Zhou Q, Li G, Du Z, Shi Y, Xing Q. ROR2 deficit may induce the tetralogy of Fallot via down-regulating of β-catenin/SOX3/HSPA6 in vitro and in vivo. J Cell Mol Med 2023; 27:3539-3552. [PMID: 37749917 PMCID: PMC10660643 DOI: 10.1111/jcmm.17969] [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: 06/18/2023] [Revised: 08/26/2023] [Accepted: 09/08/2023] [Indexed: 09/27/2023] Open
Abstract
Tetralogy of Fallot (TOF) is the highly conventional appearance of cyanotic congenital heart disease. Our study aimed to assess the involvement of receptor tyrosine kinase-like orphan receptor 2 (ROR2) in TOF and elucidate the specific mechanism. Upon investigation of human tissue samples, we observed a decrease in ROR2 expression in TOF patients compared to healthy control individuals. Transcriptome analysis revealed diminished ROR2 expression in TOF pathological samples relative to normal tissues. Of the 2246 genes that exhibited altered expression, 886 were upregulated, while 1360 were down-regulated. KEGG analysis and GO analysis of the differentially expressed genes indicated that these genes were significantly enriched in the Wnt signalling pathway, apoptosis and cardiac development function. Importantly, ROR2 was the only gene shared among the three pathways. Furthermore, interference with ROR2 promotes apoptosis and curtails cell proliferation in vitro. The knockdown of the ROR2 gene in AC16 cells resulted in a significant decrease in Edu-positive cells. Flow cytometry studies indicated an increase in the percentage of cells in the S phase. In contrast, the G2/M cell cycle transition was blocked in the ROR2-knockdown group, leading to a significant increase in apoptosis. Moreover, the CCK-8 cell viability assay demonstrated a reduced proliferation in the ROR2-knockdown group. Furthermore, both in vivo and in vitro data indicated that the expression of HSPA6 (Recombinant Heat Shock 70 kDa Protein6), an essential gene enriched in cardiac tissue and associated with apoptosis, was down-regulated following ROR2 knockdown mediated by the β-catenin/SOX3 signalling pathway. In conclusion, low expression of ROR2 plays a crucial role in the occurrence and development of TOF, which may be related to the downregulation of HSPA6 through the β-catenin/SOX3 signalling pathway.
Collapse
Affiliation(s)
- Rui Guo
- Qingdao UniversityQingdaoChina
| | | | - Qing Zhou
- The Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Guoju Li
- An Affiliated Hospital of Women and ChildrenQingdao UniversityQingdaoChina
| | - Zhanghui Du
- An Affiliated Hospital of Women and ChildrenQingdao UniversityQingdaoChina
| | - Yefei Shi
- Xuzhou Medical UniversityXuzhouChina
| | - Quansheng Xing
- An Affiliated Hospital of Women and ChildrenQingdao UniversityQingdaoChina
| |
Collapse
|
18
|
Wang J, Wan X, Le Q. Cross-regulation between SOX9 and the canonical Wnt signalling pathway in stem cells. Front Mol Biosci 2023; 10:1250530. [PMID: 37664185 PMCID: PMC10469848 DOI: 10.3389/fmolb.2023.1250530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023] Open
Abstract
SOX9, a member of the SRY-related HMG-box transcription factors, has been reported to critically regulate fetal development and stem cell homeostasis. Wnt signalling is a highly conserved signalling pathway that controls stem cell fate decision and stemness maintenance throughout embryonic development and adult life. Many studies have shown that the interactions between SOX9 and the canonical Wnt signalling pathway are involved in many of the physiological and pathological processes of stem cells, including organ development, the proliferation, differentiation and stemness maintenance of stem cells, and tumorigenesis. In this review, we summarize the already-known molecular mechanism of cross-interactions between SOX9 and the canonical Wnt signalling pathway, outline its regulatory effects on the maintenance of homeostasis in different types of stem cells, and explore its potential in translational stem cell therapy.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Xichen Wan
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Qihua Le
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Research Center, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Myopia Key Laboratory of Ministry of Health, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| |
Collapse
|
19
|
Janjanam VD, Ewart S, Zhang H, Jiang Y, Arshad H, Ziyab AH, Karmaus W. Offspring epigenetic markers at birth related to gestational BMI predict offspring BMI-trajectories from infancy to 26 years. Obes Sci Pract 2023; 9:424-434. [PMID: 37546290 PMCID: PMC10399520 DOI: 10.1002/osp4.660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 12/05/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
Objective To date, epigenetic studies identified differential DNA methylation (DNAm) related to gestational-body mass index (BMI) in offspring at birth. This study investigated whether the identified DNAm in offspring were also associated with BMI trajectories from infancy to age 26 years. Methods Data of 794 participants from Isle of Wight birth cohort in UK were investigated to study association between BMI trajectories and DNAm related to gestational-BMI at birth. Multinominal logistic regression models were applied to test the association between 1090 DNAm sites reported in three prior epigenome-wide association studies and BMI trajectories. Results DNAm site cg23089913 (NANOS1) and cg13217064 (SOX14) were associated with early persistent obesity (EPO) and delayed overweight (DOW) trajectories respectively. A higher methylation of cg23089913 showed low odds of being in EPO trajectory (OR: 0.84; 95% CI: 0.76-0.93) while higher methylation of cg13217064 resulted in 1.4-times the odds of being in DOW trajectory when compared to the normal trajectory [Correction added on 22 February 2023, after first online publication: Range of the DNAm site cg23089913 has been changed from 'lower' to 'higher' in the preceding sentence.]. In a gender-stratified analysis, the odds of developing into DOW was 1.8 times in female participants for cg13217064 while not such association was observed in males. Conclusions Deviations in methylation of cg23089913 (NANOS1) and cg13217064 (SOX14) in newborns may change the risk of having excess body weight.
Collapse
Affiliation(s)
- Vimala Devi Janjanam
- Division of Epidemiology, Biostatistics, and Environmental HealthSchool of Public HealthUniversity of MemphisMemphisTennesseeUSA
| | - Susan Ewart
- Department of Large Animal Clinical SciencesMichigan State UniversityEast LansingMichiganUSA
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics, and Environmental HealthSchool of Public HealthUniversity of MemphisMemphisTennesseeUSA
| | - Yu Jiang
- Division of Epidemiology, Biostatistics, and Environmental HealthSchool of Public HealthUniversity of MemphisMemphisTennesseeUSA
| | - Hasan Arshad
- Faculty of MedicineClinical and Experimental SciencesUniversity of SouthamptonSouthamptonUK
- The David Hide Asthma and Allergy Research CentreIsle of WightUK
- NIHR Southampton Biomedical Research CentreUniversity Hospital SouthamptonSouthamptonUK
| | - Ali H. Ziyab
- Faculty of MedicineDepartment of Community and Behavioral SciencesKuwait UniversitySafatKuwait
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics, and Environmental HealthSchool of Public HealthUniversity of MemphisMemphisTennesseeUSA
| |
Collapse
|
20
|
Cassiano LMG, Oliveira MDS, de Barros WA, de Fátima Â, Coimbra RS. Neurotoxic effects of hallucinogenic drugs 25H-NBOMe and 25H-NBOH in organotypic hippocampal cultures. Heliyon 2023; 9:e17720. [PMID: 37449113 PMCID: PMC10336585 DOI: 10.1016/j.heliyon.2023.e17720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/26/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction NBOMes and NBOHs are psychoactive drugs derived from phenethylamines and have hallucinogenic effects due to their strong agonism to serotonin 5-HT2A receptors. Although cases of toxicity associated with the recreational use of substituted phenethylamines are frequently reported, there is a lack of information on the possible neurotoxic effects of NBOMe and NBOH in the brain hippocampus, a major neurogenesis region. Objectives This study aimed at assessing the phenotypic and molecular effects of prolonged exposure of the hippocampus to the drugs 25H-NBOMe and 25H-NBOH. Methods The ex vivo organotypic culture model of hippocampal slices (OHC) was used to investigate, by immunofluorescence and confocal microscopy, and transcriptome analyses, the mechanisms associated with the neurotoxicity of 25H-NBOMe and 25H-NBOH. Results Reduction in the density of mature neurons in the OHCs occurred after two and seven days of exposure to 25H-NBOMe and 25H-NBOH, respectively. After the withdrawal of 25H-NBOMe, the density of mature neurons in the OHCs stabilized. In contrast, up to seven days after 25H-NBOH removal from the culture medium, progressive neuron loss was still observed in the OHCs. Interestingly, the exposure to 25H-NBOH induced progenitor cell differentiation, increasing the density of post-mitotic neurons in the OHCs. Corroborating these findings, the functional enrichment analysis of differentially expressed genes in the OHCs exposed to 25H-NBOH revealed the activation of WNT/Beta-catenin pathway components associated with neurogenesis. During and after the exposure to 25H-NBOMe or 25H-NBOH, gene expression patterns related to the activation of synaptic transmission and excitability of neurons were identified. Furthermore, activation of signaling pathways and biological processes related to addiction and oxidative stress and inhibition of the inflammatory response were observed after the period of drug exposure. Conclusion 25H-NBOMe and 25H-NBOH disrupt the balance between neurogenesis and neuronal death in the hippocampus and, although chemically similar, have distinct neurotoxicity mechanisms.
Collapse
Affiliation(s)
- Larissa Marcely Gomes Cassiano
- Neurogenômica, Imunopatologia, Instituto René Rachou, Fiocruz, Belo Horizonte, MG, 30190-002, Brazil
- Programa de Pós-Graduação em Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Marina da Silva Oliveira
- Neurogenômica, Imunopatologia, Instituto René Rachou, Fiocruz, Belo Horizonte, MG, 30190-002, Brazil
| | - Wellington Alves de Barros
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ângelo de Fátima
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Roney Santos Coimbra
- Neurogenômica, Imunopatologia, Instituto René Rachou, Fiocruz, Belo Horizonte, MG, 30190-002, Brazil
| |
Collapse
|
21
|
Panda M, Biswal BK. Evodiamine inhibits stemness and metastasis by altering the SOX9-β-catenin axis in non-small-cell lung cancer. J Cell Biochem 2022; 123:1454-1466. [PMID: 35788981 DOI: 10.1002/jcb.30304] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/08/2022] [Accepted: 06/22/2022] [Indexed: 01/17/2023]
Abstract
Evodiamine (EVO), a natural dietary alkaloid extracted from the roots of the Evodia rutaecarpa, has shown anticancer activities. Here, we have investigated EVO's role in inhibiting cell proliferation and migration in A549 and NCI-H522 lung cancer cells. EVO decreased the cell viability in A549 and NCI-H522 cells in a dose- and time-dependent manner. It also induced apoptosis by downregulating the expression of antiapoptotic Bcl-2 and upregulating the expression of cleaved caspase-3 and PARP. In addition, the treatment of EVO elevated the level of reactive oxygen species (ROS) generation inside the cells to induce the cell death pathways. In contrast, the pretreatment of ROS scavenger, N-acetyl cysteine, reverses the effect of EVO and attenuates cell death. Moreover, excess ROS generation in response to EVO resulted in the depletion of mitochondrial membrane potential. Furthermore, it induced DNA damage and arrested the cell cycle at the G2/M phase in A549 and NCI-H522 cells. Our study also investigated that EVO significantly suppressed tumorigenicity by inhibiting colony formation and tumorsphere formation. However, the treatment of EVO downregulated the cancer stem cell markers CD44 and CD133 in non-small-cell lung cancer. The inhibitory effect of EVO on cell invasion was mediated by altering the expression of E-cadherin, ZO-1, N-cadherin, and Vimentin. Additionally, we have revealed that EVO treatment showed downregulation of SOX9, an upstream component of β-catenin. Lastly, we have demonstrated that EVO targets the SOX9-β-catenin axis by reducing SOX9 and β-catenin expression. These findings suggested that EVO could be a promising agent for treating human lung cancer.
Collapse
Affiliation(s)
- Munmun Panda
- Department of Life Science, Cancer Drug Resistance Laboratory, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Bijesh K Biswal
- Department of Life Science, Cancer Drug Resistance Laboratory, National Institute of Technology Rourkela, Rourkela, Odisha, India
| |
Collapse
|
22
|
Mercurio S, Serra L, Pagin M, Nicolis SK. Deconstructing Sox2 Function in Brain Development and Disease. Cells 2022; 11:cells11101604. [PMID: 35626641 PMCID: PMC9139651 DOI: 10.3390/cells11101604] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
SOX2 is a transcription factor conserved throughout vertebrate evolution, whose expression marks the central nervous system from the earliest developmental stages. In humans, SOX2 mutation leads to a spectrum of CNS defects, including vision and hippocampus impairments, intellectual disability, and motor control problems. Here, we review how conditional Sox2 knockout (cKO) in mouse with different Cre recombinases leads to very diverse phenotypes in different regions of the developing and postnatal brain. Surprisingly, despite the widespread expression of Sox2 in neural stem/progenitor cells of the developing neural tube, some regions (hippocampus, ventral forebrain) appear much more vulnerable than others to Sox2 deletion. Furthermore, the stage of Sox2 deletion is also a critical determinant of the resulting defects, pointing to a stage-specificity of SOX2 function. Finally, cKOs illuminate the importance of SOX2 function in different cell types according to the different affected brain regions (neural precursors, GABAergic interneurons, glutamatergic projection neurons, Bergmann glia). We also review human genetics data regarding the brain defects identified in patients carrying mutations within human SOX2 and examine the parallels with mouse mutants. Functional genomics approaches have started to identify SOX2 molecular targets, and their relevance for SOX2 function in brain development and disease will be discussed.
Collapse
|
23
|
Target Therapy for Hepatocellular Carcinoma: Beyond Receptor Tyrosine Kinase Inhibitors and Immune Checkpoint Inhibitors. BIOLOGY 2022; 11:biology11040585. [PMID: 35453784 PMCID: PMC9027240 DOI: 10.3390/biology11040585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/19/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and its incidence is steadily increasing. The development of HCC is a complex, multi-step process that is accompanied by alterations in multiple signaling cascades. Recent years have seen advancement in understanding molecular signaling pathways that play central roles in hepatocarcinogenesis. Aberrant activation of YAP/TAZ, Hedgehog, or Wnt/β-catenin signaling is frequently found in a subset of HCC patients. Targeting the signaling pathway via small molecule inhibitors could be a promising therapeutic option for the subset of patients. In this review, we will introduce the signaling pathways, discuss their roles in the development of HCC, and propose a therapeutic approach targeting the signaling pathways in the context of HCC. Abstract Hepatocellular carcinoma (HCC) is a major health concern worldwide, and its incidence is increasing steadily. To date, receptor tyrosine kinases (RTKs) are the most favored molecular targets for the treatment of HCC, followed by immune checkpoint regulators such as PD-1, PD-L1, and CTLA-4. With less than desirable clinical outcomes from RTK inhibitors as well as immune checkpoint inhibitors (ICI) so far, novel molecular target therapies have been proposed for HCC. In this review, we will introduce diverse molecular signaling pathways that are aberrantly activated in HCC, focusing on YAP/TAZ, Hedgehog, and Wnt/β-catenin signaling pathways, and discuss potential therapeutic strategies targeting the signaling pathways in HCC.
Collapse
|
24
|
GÜNEY N, GÜRSES İ, SERİNSÖZ LİNKE E, GÜRSOY D, ÜÇBİLEK E, ÇOLAK T. Kolon adenokarsinomlarında ve adenomatöz poliplerinde Beta-Catenin ve Sox2 ekspresyonu ve klinikopatolojik parametreler ile ilişkileri. MUSTAFA KEMAL ÜNIVERSITESI TIP DERGISI 2022. [DOI: 10.17944/mkutfd.962373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Objective: Our aim was to investigate the immunohistochemical expression of β-catenin and Sox-2 in adenomatous polyps and adenocarcinoma of colon and also to evaluate the effects of these markers in adenoma-carcinoma sequence and their association with clinicopathological parameters.
Methods: Fifty-six tubular adenomas with low grade dysplasia (TALGD), 53 tubular adenomas with high grade dysplasia (TAHGD), 44 tubulovillous adenomas (TVA), 29 villous adenomas (VA) and 60 adenocarcinomas were included in the study. The nuclear staining of Sox2 was evaluated as well as both nuclear and cytoplasmic stainings of β-catenin. A semiquantitative scoring was performed. The results were compared between the groups and the relationship of the results with clinicopathological parameters was evaluated.
Results: Nuclear and cytoplasmic β-catenin expressions of the adenocarcinomas were higher than polyps. The expressions in the VA and TVA polyp groups were higher than the expressions in TAHGD and TALGD, respectively. Membranous β-catenin expression in the adenocarcinoma was higher than the polyps except VA. The evaluation between polyp groups with respect to membranous β-catenin staining revealed a statistically significantly difference in favor of VA compared with TVA, TAHGD and TALGD; in favor of TAHGD compared with TVA, in favor of TVA compared with TALGD while it was found statistically significantly higher in TAHGD than TALGD.
Conclusion: The results regarding β-catenin expression of the polyp groups were consistent with the literature. There was a positive correlation between β-catenin expression (nuclear and cytoplasmic) and malignancy. High Sox2 expressions were found correlated with malignancy potential. Large sampling size investigations to be supported by further molecular studies are needed to clarify the effect of Sox2 expression in the sequence of adenoma-carcinoma comprehensively.
Collapse
Affiliation(s)
- Neslihan GÜNEY
- UNIVERSITY OF HEALTH SCIENCES, İZMİR TEPECİK HEALTH RESEARCH CENTER, DEPARTMENT OF SURGICAL MEDICAL SCIENCES
| | - İclal GÜRSES
- İSTANBUL ÜNİVERSİTESİ-CERRAHPAŞA, CERRAHPAŞA TIP FAKÜLTESİ, CERRAHİ TIP BİLİMLERİ BÖLÜMÜ, TIBBİ PATOLOJİ ANABİLİM DALI
| | | | - Didar GÜRSOY
- HATAY MUSTAFA KEMAL ÜNİVERSİTESİ, TAYFUR ATA SÖKMEN TIP FAKÜLTESİ, CERRAHİ TIP BİLİMLERİ BÖLÜMÜ, TIBBİ PATOLOJİ ANABİLİM DALI
| | - Enver ÜÇBİLEK
- MERSIN UNIVERSITY, SCHOOL OF MEDICINE, DEPARTMENT OF INTERNAL MEDICINE, DEPARTMENT OF INTERNAL MEDICINE, GASTROENTEROLOGY
| | - Tahsin ÇOLAK
- MERSİN ÜNİVERSİTESİ, TIP FAKÜLTESİ, CERRAHİ TIP BİLİMLERİ BÖLÜMÜ, GENEL CERRAHİ ANABİLİM DALI
| |
Collapse
|
25
|
Alahdal M, Huang R, Duan L, Zhiqin D, Hongwei O, Li W, Wang D. Indoleamine 2, 3 Dioxygenase 1 Impairs Chondrogenic Differentiation of Mesenchymal Stem Cells in the Joint of Osteoarthritis Mice Model. Front Immunol 2021; 12:781185. [PMID: 34956209 PMCID: PMC8693178 DOI: 10.3389/fimmu.2021.781185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/18/2021] [Indexed: 11/29/2022] Open
Abstract
Osteoarthritis (OA) is a serious joint inflammation that leads to cartilage degeneration and joint dysfunction. Mesenchymal stem cells (MSCs) are used as a cell-based therapy that showed promising results in promoting cartilage repair. However, recent studies and clinical trials explored unsatisfied outcomes because of slow chondrogenic differentiation and increased calcification without clear reasons. Here, we report that the overexpression of indoleamine 2,3 dioxygenase 1 (IDO1) in the synovial fluid of OA patients impairs chondrogenic differentiation of MSCs in the joint of the OA mice model. The effect of MSCs mixed with IDO1 inhibitor on the cartilage regeneration was tested compared to MSCs mixed with IDO1 in the OA animal model. Further, the mechanism exploring the effect of IDO1 on chondrogenic differentiation was investigated. Subsequently, miRNA transcriptome sequencing was performed for MSCs cocultured with IDO1, and then TargetScan was used to verify the target of miR-122-5p in the SF-MSCs. Interestingly, we found that MSCs mixed with IDO1 inhibitor showed a significant performance to promote cartilage regeneration in the OA animal model, while MSCs mixed with IDO1 failed to stimulate cartilage regeneration. Importantly, the overexpression of IDO1 showed significant inhibition to Sox9 and Collagen type II (COL2A1) through activating the expression of β-catenin, since inhibiting of IDO1 significantly promoted chondrogenic signaling of MSCs (Sox9, COL2A1, Aggrecan). Further, miRNA transcriptome sequencing of SF-MSCs that treated with IDO1 showed significant downregulation of miR-122-5p which perfectly targets Wnt1. The expression of Wnt1 was noticed high when IDO1 was overexpressed. In summary, our results suggest that IDO1 overexpression in the synovial fluid of OA patients impairs chondrogenic differentiation of MSCs and cartilage regeneration through downregulation of miR-122-5p that activates the Wnt1/β-catenin pathway.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/enzymology
- Arthritis, Experimental/pathology
- Cartilage, Articular/physiology
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Chondrogenesis/drug effects
- Chondrogenesis/physiology
- Female
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/drug effects
- Mice
- MicroRNAs/metabolism
- Middle Aged
- Osteoarthritis, Knee/enzymology
- Osteoarthritis, Knee/pathology
- Rats
- Rats, Wistar
- Regeneration/drug effects
- Regeneration/physiology
- Synovial Fluid/enzymology
Collapse
Affiliation(s)
- Murad Alahdal
- Hand and Foot Surgery Department, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, China
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center), Shenzhen, China
| | - Rongxiang Huang
- Hand and Foot Surgery Department, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, China
| | - Li Duan
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center), Shenzhen, China
| | - Deng Zhiqin
- Hand and Foot Surgery Department, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, China
| | - Ouyang Hongwei
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Wencui Li
- Hand and Foot Surgery Department, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center), Shenzhen, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
26
|
Dettmer R, Niwolik I, Mehmeti I, Jörns A, Naujok O. New hPSC SOX9 and INS Reporter Cell Lines Facilitate the Observation and Optimization of Differentiation into Insulin-Producing Cells. Stem Cell Rev Rep 2021; 17:2193-2209. [PMID: 34415483 PMCID: PMC8599335 DOI: 10.1007/s12015-021-10232-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2021] [Indexed: 12/03/2022]
Abstract
Differentiation of human pluripotent stem cells into insulin-producing stem cell-derived beta cells harbors great potential for research and therapy of diabetes. SOX9 plays a crucial role during development of the pancreas and particularly in the development of insulin-producing cells as SOX9+ cells form the source for NEUROG3+ endocrine progenitor cells. For the purpose of easy monitoring of differentiation efficiencies into pancreatic progenitors and insulin-producing cells, we generated new reporter lines by knocking in a P2A-H-2Kk-F2A-GFP2 reporter gene into the SOX9-locus and a P2A-mCherry reporter gene into the INS-locus mediated by CRISPR/CAS9-technology. The knock-ins enabled co-expression of the endogenous and reporter genes and report on the endogenous gene expression. Furthermore, FACS and MACS enabled the purification of pancreatic progenitors and insulin-producing cells. Using these cell lines, we established a new differentiation protocol geared towards SOX9+ cells to efficiently drive human pluripotent stem cells into glucose-responsive beta cells. Our new protocol offers an alternative route towards stem cell-derived beta cells, pointing out the importance of Wnt/beta-catenin inhibition and the efficacy of EGF for the development of pancreatic progenitors, as well as the significance of 3D culture for the functionality of the generated beta cells.
Collapse
Affiliation(s)
- Rabea Dettmer
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Isabell Niwolik
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Ilir Mehmeti
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Ortwin Naujok
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
27
|
Sutton G, Kelsh RN, Scholpp S. Review: The Role of Wnt/β-Catenin Signalling in Neural Crest Development in Zebrafish. Front Cell Dev Biol 2021; 9:782445. [PMID: 34912811 PMCID: PMC8667473 DOI: 10.3389/fcell.2021.782445] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022] Open
Abstract
The neural crest (NC) is a multipotent cell population in vertebrate embryos with extraordinary migratory capacity. The NC is crucial for vertebrate development and forms a myriad of cell derivatives throughout the body, including pigment cells, neuronal cells of the peripheral nervous system, cardiomyocytes and skeletogenic cells in craniofacial tissue. NC induction occurs at the end of gastrulation when the multipotent population of NC progenitors emerges in the ectodermal germ layer in the neural plate border region. In the process of NC fate specification, fate-specific markers are expressed in multipotent progenitors, which subsequently adopt a specific fate. Thus, NC cells delaminate from the neural plate border and migrate extensively throughout the embryo until they differentiate into various cell derivatives. Multiple signalling pathways regulate the processes of NC induction and specification. This review explores the ongoing role of the Wnt/β-catenin signalling pathway during NC development, focusing on research undertaken in the Teleost model organism, zebrafish (Danio rerio). We discuss the function of the Wnt/β-catenin signalling pathway in inducing the NC within the neural plate border and the specification of melanocytes from the NC. The current understanding of NC development suggests a continual role of Wnt/β-catenin signalling in activating and maintaining the gene regulatory network during NC induction and pigment cell specification. We relate this to emerging models and hypotheses on NC fate restriction. Finally, we highlight the ongoing challenges facing NC research, current gaps in knowledge, and this field's potential future directions.
Collapse
Affiliation(s)
- Gemma Sutton
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Robert N. Kelsh
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
28
|
Bou-Rouphael J, Durand BC. T-Cell Factors as Transcriptional Inhibitors: Activities and Regulations in Vertebrate Head Development. Front Cell Dev Biol 2021; 9:784998. [PMID: 34901027 PMCID: PMC8651982 DOI: 10.3389/fcell.2021.784998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022] Open
Abstract
Since its first discovery in the late 90s, Wnt canonical signaling has been demonstrated to affect a large variety of neural developmental processes, including, but not limited to, embryonic axis formation, neural proliferation, fate determination, and maintenance of neural stem cells. For decades, studies have focused on the mechanisms controlling the activity of β-catenin, the sole mediator of Wnt transcriptional response. More recently, the spotlight of research is directed towards the last cascade component, the T-cell factor (TCF)/Lymphoid-Enhancer binding Factor (LEF), and more specifically, the TCF/LEF-mediated switch from transcriptional activation to repression, which in both embryonic blastomeres and mouse embryonic stem cells pushes the balance from pluri/multipotency towards differentiation. It has been long known that Groucho/Transducin-Like Enhancer of split (Gro/TLE) is the main co-repressor partner of TCF/LEF. More recently, other TCF/LEF-interacting partners have been identified, including the pro-neural BarH-Like 2 (BARHL2), which belongs to the evolutionary highly conserved family of homeodomain-containing transcription factors. This review describes the activities and regulatory modes of TCF/LEF as transcriptional repressors, with a specific focus on the functions of Barhl2 in vertebrate brain development. Specific attention is given to the transcriptional events leading to formation of the Organizer, as well as the roles and regulations of Wnt/β-catenin pathway in growth of the caudal forebrain. We present TCF/LEF activities in both embryonic and neural stem cells and discuss how alterations of this pathway could lead to tumors.
Collapse
Affiliation(s)
| | - Béatrice C. Durand
- Sorbonne Université, CNRS UMR7622, IBPS Developmental Biology Laboratory, Campus Pierre et Marie Curie, Paris, France
| |
Collapse
|
29
|
de Man SMA, Zwanenburg G, van der Wal T, Hink MA, van Amerongen R. Quantitative live-cell imaging and computational modeling shed new light on endogenous WNT/CTNNB1 signaling dynamics. eLife 2021; 10:e66440. [PMID: 34190040 PMCID: PMC8341982 DOI: 10.7554/elife.66440] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/29/2021] [Indexed: 12/16/2022] Open
Abstract
WNT/CTNNB1 signaling regulates tissue development and homeostasis in all multicellular animals, but the underlying molecular mechanism remains incompletely understood. Specifically, quantitative insight into endogenous protein behavior is missing. Here, we combine CRISPR/Cas9-mediated genome editing and quantitative live-cell microscopy to measure the dynamics, diffusion characteristics and absolute concentrations of fluorescently tagged, endogenous CTNNB1 in human cells under both physiological and oncogenic conditions. State-of-the-art imaging reveals that a substantial fraction of CTNNB1 resides in slow-diffusing cytoplasmic complexes, irrespective of the activation status of the pathway. This cytoplasmic CTNNB1 complex undergoes a major reduction in size when WNT/CTNNB1 is (hyper)activated. Based on our biophysical measurements, we build a computational model of WNT/CTNNB1 signaling. Our integrated experimental and computational approach reveals that WNT pathway activation regulates the dynamic distribution of free and complexed CTNNB1 across different subcellular compartments through three regulatory nodes: the destruction complex, nucleocytoplasmic shuttling, and nuclear retention.
Collapse
Affiliation(s)
- Saskia MA de Man
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Gooitzen Zwanenburg
- Biosystems Data Analysis, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Tanne van der Wal
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Mark A Hink
- Molecular Cytology, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
- van Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| |
Collapse
|
30
|
Iwata T, Mizuno N, Ishida S, Kajiya M, Nagahara T, Kaneda-Ikeda E, Yoshioka M, Munenaga S, Ouhara K, Fujita T, Kawaguchi H, Kurihara H. Functional Regulatory Mechanisms Underlying Bone Marrow Mesenchymal Stem Cell Senescence During Cell Passages. Cell Biochem Biophys 2021; 79:321-336. [PMID: 33559812 DOI: 10.1007/s12013-021-00969-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cell (MSC) transplantation is an effective periodontal regenerative therapy. MSCs are multipotent, have self-renewal ability, and can differentiate into periodontal cells. However, senescence is inevitable for MSCs. In vitro, cell senescence can be induced by long-term culture with/without cell passage. However, the regulatory mechanism of MSC senescence remains unclear. Undifferentiated MSC-specific transcription factors can regulate MSC function. Herein, we identified the regulatory transcription factors involved in MSC senescence and elucidated their mechanisms of action. We cultured human MSCs (hMSCs) with repetitive cell passages to induce cell senescence and evaluated the mRNA and protein expression of cell senescence-related genes. Additionally, we silenced the cell senescence-induced transcription factors, GATA binding protein 6 (GATA6) and SRY-box 11 (SOX11), and investigated senescence-related signaling pathways. With repeated passages, the number of senescent cells increased, while the cell proliferation capacity decreased; GATA6 mRNA expression was upregulated and that of SOX11 was downregulated. Repetitive cell passages decreased Wnt and bone morphogenetic protein (BMP) signaling pathway-related gene expression. Silencing of GATA6 and SOX11 regulated Wnt and BMP signaling pathway-related genes and affected cell senescence-related genes; moreover, SOX11 silencing regulated GATA6 expression. Hence, we identified them as pair of regulatory transcription factors for cell senescence in hMSCs via the Wnt and BMP signaling pathways.
Collapse
Affiliation(s)
- T Iwata
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan.
| | - N Mizuno
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - S Ishida
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - M Kajiya
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - T Nagahara
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - E Kaneda-Ikeda
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - M Yoshioka
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - S Munenaga
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
- Department of General Dentistry, Hiroshima University Hospital, Hiroshima, 734-8553, Japan
| | - K Ouhara
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - T Fujita
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - H Kawaguchi
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
- Department of General Dentistry, Hiroshima University Hospital, Hiroshima, 734-8553, Japan
| | - H Kurihara
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| |
Collapse
|
31
|
de Moraes GFA, Listik E, Justo GZ, Vicente CM, Toma L. The Glypican proteoglycans show intrinsic interactions with Wnt-3a in human prostate cancer cells that are not always associated with cascade activation. BMC Mol Cell Biol 2021; 22:26. [PMID: 33947326 PMCID: PMC8097805 DOI: 10.1186/s12860-021-00361-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 04/01/2021] [Indexed: 01/12/2023] Open
Abstract
Background Prostate cancer occurs through multiple steps until advanced metastasis. Signaling pathways studies can result in the identification of targets to interrupt cancer progression. Glypicans are cell surface proteoglycans linked to the membrane through glycosylphosphatidylinositol. Their interaction with specific ligands has been reported to trigger diverse signaling, including Wnt. In this study, prostate cancer cell lines PC-3, DU-145, and LNCaP were compared to normal prostate RWPE-1 cell line to investigate glypican family members and the activation of the Wnt signaling pathway. Results Glypican-1 (GPC1) was highly expressed in all the examined cell lines, except for LNCaP, which expressed glypican-5 (GPC5). The subcellular localization of GPC1 was detected on the cell surface of RWPE-1, PC-3, and DU-145 cell lines, while GPC5 suggested cytoplasm localization in LNCaP cells. Besides glypican, flow cytometry analysis in these prostate cell lines confirmed the expression of Wnt-3a and unphosphorylated β-catenin. The co-immunoprecipitation assay revealed increased levels of binding between Wnt-3a and glypicans in cancer cells, suggesting a relationship between these proteoglycans in this pathway. A marked increase in nuclear β-catenin was observed in tumor cells. However, only PC-3 cells demonstrated activation of canonical Wnt signaling, according to the TOPFLASH assay. Conclusions GPC1 was the majorly expressed gene in all the studied cell lines, except for LNCaP, which expressed GPC5. We assessed by co-immunoprecipitation that these GPCs could interact with Wnt-3a. However, even though nuclear β-catenin was found increased in the prostate cancer cells (i.e., PC-3, DU-145 and LNCaP), activation of Wnt pathway was only found in PC-3 cells. In these PC-3 cells, GPC1 and Wnt-3a revealed high levels of colocalization, as assessed by confocal microscopy studies. This suggests a localization at the cellular surface, where Frizzled receptor is required for downstream activation. The interaction of Wnt-3a with GPCs in DU-145 and LNCaP cells, which occurs in absence of Wnt signaling activation, requires further studies. Once non-TCF-LEF proteins can also bind β-catenin, another signaling pathway may be involved in these cells with regulatory function. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00361-x.
Collapse
Affiliation(s)
- Gabrielle Ferrante Alves de Moraes
- Departamento de Bioquímica (Campus São Paulo), Universidade Federal de São Paulo, Rua Três de Maio, P.O. Box: 04044-020, São Paulo, SP, 100, Brazil
| | - Eduardo Listik
- Departamento de Bioquímica (Campus São Paulo), Universidade Federal de São Paulo, Rua Três de Maio, P.O. Box: 04044-020, São Paulo, SP, 100, Brazil
| | - Giselle Zenker Justo
- Departamento de Bioquímica (Campus São Paulo), Universidade Federal de São Paulo, Rua Três de Maio, P.O. Box: 04044-020, São Paulo, SP, 100, Brazil.,Departamento de Ciências Biológicas (Campus Diadema), Universidade Federal de São Paulo, Rua Três de Maio, P.O. Box: 04044-020, São Paulo, SP, 100, Brazil
| | - Carolina Meloni Vicente
- Departamento de Bioquímica (Campus São Paulo), Universidade Federal de São Paulo, Rua Três de Maio, P.O. Box: 04044-020, São Paulo, SP, 100, Brazil
| | - Leny Toma
- Departamento de Bioquímica (Campus São Paulo), Universidade Federal de São Paulo, Rua Três de Maio, P.O. Box: 04044-020, São Paulo, SP, 100, Brazil.
| |
Collapse
|
32
|
Stevanovic M, Drakulic D, Lazic A, Ninkovic DS, Schwirtlich M, Mojsin M. SOX Transcription Factors as Important Regulators of Neuronal and Glial Differentiation During Nervous System Development and Adult Neurogenesis. Front Mol Neurosci 2021; 14:654031. [PMID: 33867936 PMCID: PMC8044450 DOI: 10.3389/fnmol.2021.654031] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
The SOX proteins belong to the superfamily of transcription factors (TFs) that display properties of both classical TFs and architectural components of chromatin. Since the cloning of the Sox/SOX genes, remarkable progress has been made in illuminating their roles as key players in the regulation of multiple developmental and physiological processes. SOX TFs govern diverse cellular processes during development, such as maintaining the pluripotency of stem cells, cell proliferation, cell fate decisions/germ layer formation as well as terminal cell differentiation into tissues and organs. However, their roles are not limited to development since SOX proteins influence survival, regeneration, cell death and control homeostasis in adult tissues. This review summarized current knowledge of the roles of SOX proteins in control of central nervous system development. Some SOX TFs suspend neural progenitors in proliferative, stem-like state and prevent their differentiation. SOX proteins function as pioneer factors that occupy silenced target genes and keep them in a poised state for activation at subsequent stages of differentiation. At appropriate stage of development, SOX members that maintain stemness are down-regulated in cells that are competent to differentiate, while other SOX members take over their functions and govern the process of differentiation. Distinct SOX members determine down-stream processes of neuronal and glial differentiation. Thus, sequentially acting SOX TFs orchestrate neural lineage development defining neuronal and glial phenotypes. In line with their crucial roles in the nervous system development, deregulation of specific SOX proteins activities is associated with neurodevelopmental disorders (NDDs). The overview of the current knowledge about the link between SOX gene variants and NDDs is presented. We outline the roles of SOX TFs in adult neurogenesis and brain homeostasis and discuss whether impaired adult neurogenesis, detected in neurodegenerative diseases, could be associated with deregulation of SOX proteins activities. We present the current data regarding the interaction between SOX proteins and signaling pathways and microRNAs that play roles in nervous system development. Finally, future research directions that will improve the knowledge about distinct and various roles of SOX TFs in health and diseases are presented and discussed.
Collapse
Affiliation(s)
- Milena Stevanovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia.,Faculty of Biology, University of Belgrade, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Danijela Drakulic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Andrijana Lazic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Danijela Stanisavljevic Ninkovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marija Schwirtlich
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marija Mojsin
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
33
|
Barros II, Leão V, Santis JO, Rosa RCA, Brotto DB, Storti CB, Siena ÁDD, Molfetta GA, Silva WA. Non-Syndromic Intellectual Disability and Its Pathways: A Long Noncoding RNA Perspective. Noncoding RNA 2021; 7:ncrna7010022. [PMID: 33799572 PMCID: PMC8005948 DOI: 10.3390/ncrna7010022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Non-syndromic intellectual disability (NS-ID or idiopathic) is a complex neurodevelopmental disorder that represents a global health issue. Although many efforts have been made to characterize it and distinguish it from syndromic intellectual disability (S-ID), the highly heterogeneous aspect of this disorder makes it difficult to understand its etiology. Long noncoding RNAs (lncRNAs) comprise a large group of transcripts that can act through various mechanisms and be involved in important neurodevelopmental processes. In this sense, comprehending the roles they play in this intricate context is a valuable way of getting new insights about how NS-ID can arise and develop. In this review, we attempt to bring together knowledge available in the literature about lncRNAs involved with molecular and cellular pathways already described in intellectual disability and neural function, to better understand their relevance in NS-ID and the regulatory complexity of this disorder.
Collapse
Affiliation(s)
- Isabela I. Barros
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Vitor Leão
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Jessica O. Santis
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Reginaldo C. A. Rosa
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Danielle B. Brotto
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Camila B. Storti
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Ádamo D. D. Siena
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Greice A. Molfetta
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Wilson A. Silva
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
- National Institute of Science and Technology in Stem Cell and Cell Therapy and Center for Cell Based Therapy, Ribeirão Preto Medical School, University of São Paulo, Rua Tenente Catão Roxo, 2501, Monte Alegre, Ribeirão Preto 14051-140, Brazil
- Center for Integrative Systems Biology-CISBi, NAP/USP, Ribeirão Preto Medical School, University of São Paulo, Rua Catão Roxo, 2501, Monte Alegre, Ribeirão Preto 14051-140, Brazil
- Department of Medicine at the Midwest State University of Paraná-UNICENTRO, and Guarapuava Institute for Cancer Research, Rua Fortim Atalaia, 1900, Cidade dos Lagos, Guarapuava 85100-000, Brazil
- Correspondence: ; Tel.: +55-16-3315-3293
| |
Collapse
|
34
|
Hirate Y, Hayakawa K, Nakano Y, Kumazawa S, Miura K, Kanai Y, Kanai-Azuma M. Early Crypt Formation Defects in the Uterine Epithelia of Sox17 Heterozygous Mice. Sex Dev 2021; 14:40-50. [PMID: 33690235 DOI: 10.1159/000513386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/27/2020] [Indexed: 11/19/2022] Open
Abstract
SOX17 activity in the uterine epithelium is essential for the implantation of mouse embryos. Previously, we demonstrated that female Sox17 heterozygous mutant mice are subfertile, and 2 active copies of Sox17 are required for the proper implantation of mouse embryos. To understand which implantation step is most sensitive to the Sox17 gene dosage, we comprehensively investigated the phenotypes and RNA transcriptomes of Sox17 heterozygous mutant mice. Uterine Sox17 expression drastically changed according to estrous cycle and during early pregnancy. The highest Sox17 expression was observed during the receptive period for blastocyst implantation. Sox17 heterozygous uterine epithelia showed ectopic high-level expression of SOX9, another SOX factor that is normally expressed in the uterine gland. Three-dimensional analysis of the uterus on day 5 of pregnancy revealed no crypt formation near the healthy blastocysts in the Sox17 heterozygous uterine epithelium, suggesting that early defects in embryo homing had occurred. Global transcriptional analysis revealed that the expression of Amphiregulin (Areg), a gene encoding a heparin-binding epidermal growth factor receptor ligand, was decreased drastically in Sox17+/- uterine epithelia. These data imply that full Sox17 activity is required to promote early crypt formation through proper regulation of SOX9 and AREG expression at the implantation site.
Collapse
Affiliation(s)
- Yoshikazu Hirate
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Center for Experimental Animals, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kana Hayakawa
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuki Nakano
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shiori Kumazawa
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kento Miura
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
| | - Yoshiakira Kanai
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
| | - Masami Kanai-Azuma
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan, .,Center for Experimental Animals, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,
| |
Collapse
|
35
|
Wu Y, Wharton J, Walters R, Vasilaki E, Aman J, Zhao L, Wilkins MR, Rhodes CJ. The pathophysiological role of novel pulmonary arterial hypertension gene SOX17. Eur Respir J 2021; 58:13993003.04172-2020. [PMID: 33632800 DOI: 10.1183/13993003.04172-2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/08/2021] [Indexed: 11/05/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease predominantly targeting pre-capillary blood vessels. Adverse structural remodelling and increased pulmonary vascular resistance result in cardiac hypertrophy and ultimately failure of the right ventricle. Recent whole-genome and whole-exome sequencing studies have identified SOX17 as a novel risk gene in PAH, with a dominant mode of inheritance and incomplete penetrance. Rare deleterious variants in the gene and more common variants in upstream enhancer sites have both been associated with the disease, and a deficiency of SOX17 expression may predispose to PAH. This review aims to consolidate the evidence linking genetic variants in SOX17 to PAH, and explores the numerous targets and effects of the transcription factor, focusing on the pulmonary vasculature and the pathobiology of PAH.
Collapse
Affiliation(s)
- Yukyee Wu
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Rachel Walters
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Eleni Vasilaki
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jurjan Aman
- National Heart and Lung Institute, Imperial College London, London, UK.,VUmc, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lan Zhao
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
36
|
Bantle CM, French CT, Cummings JE, Sadasivan S, Tran K, Slayden RA, Smeyne RJ, Tjalkens RB. Manganese exposure in juvenile C57BL/6 mice increases glial inflammatory responses in the substantia nigra following infection with H1N1 influenza virus. PLoS One 2021; 16:e0245171. [PMID: 33493177 PMCID: PMC7833173 DOI: 10.1371/journal.pone.0245171] [Citation(s) in RCA: 4] [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: 11/04/2020] [Accepted: 12/22/2020] [Indexed: 01/22/2023] Open
Abstract
Infection with Influenza A virus can lead to the development of encephalitis and subsequent neurological deficits ranging from headaches to neurodegeneration. Post-encephalitic parkinsonism has been reported in surviving patients of H1N1 infections, but not all cases of encephalitic H1N1 infection present with these neurological symptoms, suggesting that interactions with an environmental neurotoxin could promote more severe neurological damage. The heavy metal, manganese (Mn), is a potential interacting factor with H1N1 because excessive exposure early in life can induce long-lasting effects on neurological function through inflammatory activation of glial cells. In the current study, we used a two-hit model of neurotoxin-pathogen exposure to examine whether exposure to Mn during juvenile development would induce a more severe neuropathological response following infection with H1N1 in adulthood. To test this hypothesis, C57BL/6 mice were exposed to MnCl2 in drinking water (50 mg/kg/day) for 30 days from days 21–51 postnatal, then infected intranasally with H1N1 three weeks later. Analyses of dopaminergic neurons, microglia and astrocytes in basal ganglia indicated that although there was no significant loss of dopaminergic neurons within the substantia nigra pars compacta, there was more pronounced activation of microglia and astrocytes in animals sequentially exposed to Mn and H1N1, as well as altered patterns of histone acetylation. Whole transcriptome Next Generation Sequencing (RNASeq) analysis was performed on the substantia nigra and revealed unique patterns of gene expression in the dual-exposed group, including genes involved in antioxidant activation, mitophagy and neurodegeneration. Taken together, these results suggest that exposure to elevated levels of Mn during juvenile development could sensitize glial cells to more severe neuro-immune responses to influenza infection later in life through persistent epigenetic changes.
Collapse
Affiliation(s)
- Collin M. Bantle
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - C. Tenley French
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jason E. Cummings
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Shankar Sadasivan
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Kevin Tran
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Richard A. Slayden
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Richard J. Smeyne
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Ronald B. Tjalkens
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
37
|
Wang J, Ahimaz PR, Hashemifar S, Khlevner J, Picoraro JA, Middlesworth W, Elfiky MM, Que J, Shen Y, Chung WK. Novel candidate genes in esophageal atresia/tracheoesophageal fistula identified by exome sequencing. Eur J Hum Genet 2021; 29:122-130. [PMID: 32641753 PMCID: PMC7852873 DOI: 10.1038/s41431-020-0680-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/07/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
The various malformations of the aerodigestive tract collectively known as esophageal atresia/tracheoesophageal fistula (EA/TEF) constitute a rare group of birth defects of largely unknown etiology. Previous studies have identified a small number of rare genetic variants causing syndromes associated with EA/TEF. We performed a pilot exome sequencing study of 45 unrelated simplex trios (probands and parents) with EA/TEF. Thirteen had isolated and 32 had nonisolated EA/TEF; none had a family history of EA/TEF. We identified de novo variants in protein-coding regions, including 19 missense variants predicted to be deleterious (D-mis) and 3 likely gene-disrupting (LGD) variants. Consistent with previous studies of structural birth defects, there is a trend of increased burden of de novo D-mis in cases (1.57-fold increase over the background mutation rate), and the burden is greater in constrained genes (2.55-fold, p = 0.003). There is a frameshift de novo variant in EFTUD2, a known EA/TEF risk gene involved in mRNA splicing. Strikingly, 15 out of 19 de novo D-mis variants are located in genes that are putative target genes of EFTUD2 or SOX2 (another known EA/TEF gene), much greater than expected by chance (3.34-fold, p value = 7.20e-5). We estimated that 33% of patients can be attributed to de novo deleterious variants in known and novel genes. We identified APC2, AMER3, PCDH1, GTF3C1, POLR2B, RAB3GAP2, and ITSN1 as plausible candidate genes in the etiology of EA/TEF. We conclude that further genomic analysis to identify de novo variants will likely identify previously undescribed genetic causes of EA/TEF.
Collapse
Affiliation(s)
- Jiayao Wang
- grid.239585.00000 0001 2285 2675Department of Pediatrics, Columbia University Medical Center, New York, NY USA ,grid.239585.00000 0001 2285 2675Departments of Systems Biology and Biomedical Informatics, Columbia University Medical Center, New York, NY USA
| | - Priyanka R. Ahimaz
- grid.239585.00000 0001 2285 2675Department of Pediatrics, Columbia University Medical Center, New York, NY USA
| | - Somaye Hashemifar
- grid.239585.00000 0001 2285 2675Department of Pediatrics, Columbia University Medical Center, New York, NY USA ,grid.239585.00000 0001 2285 2675Departments of Systems Biology and Biomedical Informatics, Columbia University Medical Center, New York, NY USA
| | - Julie Khlevner
- grid.239585.00000 0001 2285 2675Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Columbia University Medical Center, New York, NY USA
| | - Joseph A. Picoraro
- grid.239585.00000 0001 2285 2675Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Columbia University Medical Center, New York, NY USA
| | - William Middlesworth
- grid.239585.00000 0001 2285 2675Division of Pediatric Surgery, Department of Surgery, Columbia University Medical Center, New York, NY USA
| | - Mahmoud M. Elfiky
- grid.7776.10000 0004 0639 9286Pediatric Surgery, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Jianwen Que
- grid.239585.00000 0001 2285 2675Department of Medicine, Columbia University Medical Center, New York, NY USA
| | - Yufeng Shen
- grid.239585.00000 0001 2285 2675Departments of Systems Biology and Biomedical Informatics, Columbia University Medical Center, New York, NY USA
| | - Wendy K. Chung
- grid.239585.00000 0001 2285 2675Department of Pediatrics, Columbia University Medical Center, New York, NY USA ,grid.239585.00000 0001 2285 2675Department of Medicine, Columbia University Medical Center, New York, NY USA
| |
Collapse
|
38
|
Tian W, Li Z, Bai L, Chen L, Yan Y, Li H, Han Y, Teng F, Gao C, Xue F, Wang Y. The oncogenic role of SOX8 in endometrial carcinoma. Cancer Biol Ther 2020; 21:1136-1144. [PMID: 33190587 DOI: 10.1080/15384047.2020.1840318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Endometrial carcinoma (EC) remains one of the most prevalent forms of cancer to impact the female reproductive system, yet the mechanisms governing its development and progression are incompletely understood. We, therefore, sought to assess the relevance of SOX8 to EC progression and patient prognosis. Array comparative genomic hybridization (aCGH) was performed using samples from 50 patients with EC. Samples were separated based upon whether patients were positive for lymph node metastasis (LN+ and LN-, respectively). Based on our initial results, the SOX8 gene was selected for further analysis. Immunohistochemical staining of 630 endometrial tissue samples was conducted to understand how SOX8 expression relates to specific EC clinicopathological characteristics. In addition, we explored the impact of SOX8 expression on the growth, invasion, and migration of EC cells through knockdown and overexpression experiments. In our initial aCGH analysis, SOX family proteins and the Wnt and Notch signaling pathways were significantly associated with EC LN metastasis. SOX8 expression was markedly increased in EC tumor samples relative to normal endometrial tissue (P= .003), and higher SOX8 expression was linked to a high tumor histological grade (P= .032), LN metastasis (P= .027), and shorter patient overall survival (P= .031). When SOX8 was knocked down, this further impaired the proliferative, invasive, and migratory activity of EC cells, whereas overexpressing this gene had the opposite effect. SOX8 may function in an oncogenic manner to drive EC development and progression, and higher SOX8 expression is associated with a poor EC patient prognosis.
Collapse
Affiliation(s)
- Wenyan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China
| | - Zhanghuan Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China
| | - Lu Bai
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China.,Department of Gynecology and Obstetrics, Nankai University Affiliated Hospital (Tianjin No.4 Hospital) , Tianjin, China
| | - Lingli Chen
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China
| | - Ye Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China
| | - Huihui Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China
| | - Yanyan Han
- Department of Pathology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine , Tianjin, China
| | - Fei Teng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China
| | - Chao Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital , Tianjin, China
| |
Collapse
|
39
|
Ahmad A, Strohbuecker S, Scotti C, Tufarelli C, Sottile V. In Silico Identification of SOX1 Post-Translational Modifications Highlights a Shared Protein Motif. Cells 2020; 9:E2471. [PMID: 33202879 PMCID: PMC7696889 DOI: 10.3390/cells9112471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/10/2020] [Indexed: 12/02/2022] Open
Abstract
The transcription factor SOX1 is a key regulator of neural stem cell development, acting to keep neural stem cells (NSCs) in an undifferentiated state. Postnatal expression of Sox1 is typically confined to the central nervous system (CNS), however, its expression in non-neural tissues has recently been implicated in tumorigenesis. The mechanism through which SOX1 may exert its function is not fully understood, and studies have mainly focused on changes in SOX1 expression at a transcriptional level, while its post-translational regulation remains undetermined. To investigate this, data were extracted from different publicly available databases and analysed to search for putative SOX1 post-translational modifications (PTMs). Results were compared to PTMs associated with SOX2 in order to identify potentially key PTM motifs common to these SOXB1 proteins, and mapped on SOX1 domain structural models. This approach identified several putative acetylation, phosphorylation, glycosylation and sumoylation sites within known functional domains of SOX1. In particular, a novel SOXB1 motif (xKSExSxxP) was identified within the SOX1 protein, which was also found in other unrelated proteins, most of which were transcription factors. These results also highlighted potential phospho-sumoyl switches within this SOXB1 motif identified in SOX1, which could regulate its transcriptional activity. This analysis indicates different types of PTMs within SOX1, which may influence its regulatory role as a transcription factor, by bringing changes to its DNA binding capacities and its interactions with partner proteins. These results provide new research avenues for future investigations on the mechanisms regulating SOX1 activity, which could inform its roles in the contexts of neural stem cell development and cancer.
Collapse
Affiliation(s)
- Azaz Ahmad
- School of Medicine, The University of Nottingham, Nottingham NG7 2RD, UK; (A.A.); (S.S.)
| | - Stephanie Strohbuecker
- School of Medicine, The University of Nottingham, Nottingham NG7 2RD, UK; (A.A.); (S.S.)
| | - Claudia Scotti
- Department of Molecular Medicine, The University of Pavia, 27100 Pavia, Italy;
| | - Cristina Tufarelli
- Department of Genetics and Genome Biology/Leicester Cancer Research Centre, The University of Leicester, Leicester LE2 7LX, UK;
| | - Virginie Sottile
- School of Medicine, The University of Nottingham, Nottingham NG7 2RD, UK; (A.A.); (S.S.)
- Department of Molecular Medicine, The University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
40
|
Mukherjee S, Chaturvedi P, Rankin SA, Fish MB, Wlizla M, Paraiso KD, MacDonald M, Chen X, Weirauch MT, Blitz IL, Cho KW, Zorn AM. Sox17 and β-catenin co-occupy Wnt-responsive enhancers to govern the endoderm gene regulatory network. eLife 2020; 9:58029. [PMID: 32894225 PMCID: PMC7498262 DOI: 10.7554/elife.58029] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/04/2020] [Indexed: 12/30/2022] Open
Abstract
Lineage specification is governed by gene regulatory networks (GRNs) that integrate the activity of signaling effectors and transcription factors (TFs) on enhancers. Sox17 is a key transcriptional regulator of definitive endoderm development, and yet, its genomic targets remain largely uncharacterized. Here, using genomic approaches and epistasis experiments, we define the Sox17-governed endoderm GRN in Xenopus gastrulae. We show that Sox17 functionally interacts with the canonical Wnt pathway to specify and pattern the endoderm while repressing alternative mesectoderm fates. Sox17 and β-catenin co-occupy hundreds of key enhancers. In some cases, Sox17 and β-catenin synergistically activate transcription apparently independent of Tcfs, whereas on other enhancers, Sox17 represses β-catenin/Tcf-mediated transcription to spatially restrict gene expression domains. Our findings establish Sox17 as a tissue-specific modifier of Wnt responses and point to a novel paradigm where genomic specificity of Wnt/β-catenin transcription is determined through functional interactions between lineage-specific Sox TFs and β-catenin/Tcf transcriptional complexes. Given the ubiquitous nature of Sox TFs and Wnt signaling, this mechanism has important implications across a diverse range of developmental and disease contexts.
Collapse
Affiliation(s)
- Shreyasi Mukherjee
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, United States
| | - Praneet Chaturvedi
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, United States
| | - Scott A Rankin
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, United States
| | - Margaret B Fish
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, United States
| | - Marcin Wlizla
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Kitt D Paraiso
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, United States.,Center for Complex Biological Systems, University of California, Irvine, Irvine, United States
| | - Melissa MacDonald
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, United States
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology (CAGE), Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Matthew T Weirauch
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, United States.,Center for Autoimmune Genomics and Etiology (CAGE), Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Ira L Blitz
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, United States
| | - Ken Wy Cho
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, United States
| | - Aaron M Zorn
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, United States
| |
Collapse
|
41
|
Lozano E, Asensio M, Perez-Silva L, Banales JM, Briz O, Marin JJG. MRP3-Mediated Chemoresistance in Cholangiocarcinoma: Target for Chemosensitization Through Restoring SOX17 Expression. Hepatology 2020; 72:949-964. [PMID: 31863486 DOI: 10.1002/hep.31088] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 12/10/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND AIMS A limitation for the treatment of unresectable cholangiocarcinoma (CCA) is its poor response to chemotherapy, which is partly due to reduction of intracellular levels of anticancer drugs through ATP-binding cassette (ABC) pumps. Low expression of SOX17 (SRY-box containing gene 17), a transcription factor that promotes biliary differentiation and phenotype maintenance, has been associated with cholangiocyte malignant transformation. Whether SOX17 is also involved in CCA chemoresistance is investigated in this study. APPROACH AND RESULTS SOX17 expression in human CCA cells (EGI-1 and TFK-1) selectively potentiated cytotoxicity of SN-38, 5-fluorouracil and mitoxantrone, but not that of gemcitabine, capecitabine, cisplatin, or oxaliplatin. The analysis of the resistome by TaqMan low-density arrays revealed changes affecting primarily ABC pump expression. Single-gene quantitative real-time PCR, immunoblot, and immunofluorescence analyses confirmed that MRP3 (multidrug resistance associated protein 3), which was highly expressed in CCA human tumors, was down-regulated in SOX17-transduced CCA cells. The substrate specificity of this pump matched that of SOX17-induced in vitro selective chemosensitization. Functional studies showed lower ability of SOX17-expressing CCA cells to extrude specific MRP3 substrates. Reporter assay of MRP3 promoter (ABCC3pr) revealed that ABCC3pr activity was inhibited by SOX17 expression and SOX2/SOX9 silencing. The latter was highly expressed in CCA. Moreover, SOX2/9, but not SOX17, induced altered electrophoretic mobility of ABCC3pr, which was prevented by SOX17. The growth of CCA tumors subcutaneously implanted into immunodeficient mice was inhibited by 5-fluorouracil. This effect was enhanced by co-treatment with adenoviral vectors encoding SOX17. CONCLUSIONS SOX9/2/17 are involved in MRP3-mediated CCA chemoresistance. Restored SOX17 expression, in addition to its tumor suppression effect, induces selective chemosensitization due to MRP3 down-regulation and subsequent intracellular drug accumulation.
Collapse
Affiliation(s)
- Elisa Lozano
- Experimental Hepatology and Drug Targeting, IBSAL, University of Salamanca, Salamanca, Spain.,National Institute for the Study of Liver and Gastrointestinal Diseases, Carlos III National Health Institute, Madrid, Spain
| | - Maitane Asensio
- Experimental Hepatology and Drug Targeting, IBSAL, University of Salamanca, Salamanca, Spain
| | - Laura Perez-Silva
- Experimental Hepatology and Drug Targeting, IBSAL, University of Salamanca, Salamanca, Spain
| | - Jesus M Banales
- National Institute for the Study of Liver and Gastrointestinal Diseases, Carlos III National Health Institute, Madrid, Spain.,Department of Hepatology and Gastroenterology, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country, San Sebastian, Spain.,Ikerbasque, Bilbao, Spain
| | - Oscar Briz
- Experimental Hepatology and Drug Targeting, IBSAL, University of Salamanca, Salamanca, Spain.,National Institute for the Study of Liver and Gastrointestinal Diseases, Carlos III National Health Institute, Madrid, Spain
| | - Jose J G Marin
- Experimental Hepatology and Drug Targeting, IBSAL, University of Salamanca, Salamanca, Spain.,National Institute for the Study of Liver and Gastrointestinal Diseases, Carlos III National Health Institute, Madrid, Spain
| |
Collapse
|
42
|
Huang J, Gao H, Tan HZ. SOX1 Promoter Hypermethylation as a Potential Biomarker for High-Grade Squamous Intraepithelial Neoplasia Lesion and Cervical Carcinoma: A Meta-Analysis With Trial Sequential Analysis. Front Genet 2020; 11:633. [PMID: 32849763 PMCID: PMC7411256 DOI: 10.3389/fgene.2020.00633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
Background: DNA methylation has been widely assessed as a potential biomarker for the early detection of cervical cancer (CC). Herein, we assessed the associations of SOX1 promoter hypermethylation with squamous intraepithelial lesion and CC. Methods: Published studies and genome-wide methylation datasets were searched from electronic databases (up to April 2019). The associations of SOX1 hypermethylation with high-grade squamous intraepithelial lesion (HSIL) and CC risks were evaluated by odds ratios (ORs) and 95% confidence intervals (CIs). The summary receiver operator characteristic test was used to assess the diagnostic value of the SOX1 promoter hypermethylation of CC and intraepithelial neoplasia type III or worse (CIN3+). Trial sequential analysis (TSA) was performed to evaluate the stability of results and estimate the required information size (RIS). Results: In this meta-analysis of 17 published studies, the SOX1 methylation rates increased among low-grade SIL (LSIL, 27.27%), HSIL (40.75%), and CC (84.56%) specimens. Compared with control specimens, SOX1 promoter hypermethylation progressively increased the risk of HSIL by 4.20-fold (p < 0.001) and CC by 41.26-fold (p < 0.001). The pooled sensitivity of SOX1 methylation was estimated to be 0.85 (95% CI: 0.81–0.88) in differentiating patients with CC, corresponding to a specificity of 0.72 (95% CI: 0.69–0.75) and an area under the curve (AUC) of 0.93. Furthermore, the pooled sensitivity of SOX1 methylation was estimated to be 0.75 (95% CI: 0.72–0.78) in differentiating patients with CIN3+, corresponding to a specificity of 0.71 (95% CI: 0.69–0.73) and an AUC of 0.84. The pooled results of TCGA and GEO datasets showed that all CpG sites in SOX1 were associated with CC and 16 of 19 CpG sites were associated with HSIL. The results of TSA illustrated that the size was sufficient and significant associations were observed. Conclusion: This meta-analysis indicated that SOX1 promoter hypermethylation might have a potential value in the clinical diagnosis of CC and CIN3+.
Collapse
Affiliation(s)
- Jin Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, China
| | - Hong Gao
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, China.,School of Nursing, University of South China, Hengyang, China
| | - Hong-Zhuan Tan
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, China
| |
Collapse
|
43
|
Chen L, Xie Y, Ma X, Zhang Y, Li X, Zhang F, Gao Y, Fan Y, Gu L, Wang L, Zhang X, Fu B. SOX6 represses tumor growth of clear cell renal cell carcinoma by HMG domain-dependent regulation of Wnt/β-catenin signaling. Mol Carcinog 2020; 59:1159-1173. [PMID: 32794610 DOI: 10.1002/mc.23246] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/26/2020] [Accepted: 08/05/2020] [Indexed: 01/18/2023]
Abstract
Sex-determining region Y box (SOXs) are expressed in various cells and control cell fate and differentiation in a multitude of physiologic processes. SOX6, a main representative of SOXs, is involved in the regulation of carcinogenesis in various human malignancies. However, the role of SOX6 in clear cell renal cell carcinoma (ccRCC) remains unclear. In this study, SOX6 expression in ccRCC and its clinical significance were investigated. In vitro and in vivo assays were used to explore the tumor-related function and the underlying molecular mechanism of SOX6 in ccRCC. We confirmed that SOX6 was frequently downregulated in ccRCC tissues and cell lines. Besides, downregulation of SOX6 was significantly associated with larger tumor sizes, advanced tumor stage, higher Fuhrman grades, and its expression could act as an independent prognostic factor for ccRCC (hazards ratio = 0.590, P = .026). Gain/loss-of-function experiments demonstrated that SOX6 could remarkably inhibit tumor cell growth and foci formation in vitro and xenograft tumorigenesis in vivo, respectively. Mechanistically, SOX6 could influence cell cycle by regulating the G1/the S phase transition and had an inhibitory effect on Wnt/β-catenin signaling as well as its target genes, c-Myc and cyclin D1. Interesting, the tumor-suppressive function of SOX6 was proved to be dependent on its specific high-mobility-group (HMG) domain. In general, our findings indicated that SOX6 was a novel tumor suppressor and prognostic biomarker in ccRCC. SOX6 could inhibit tumor growth by negatively regulating the Wnt/β-catenin signaling pathway in an HMG domain-dependent manner in ccRCC, which might provide a novel therapeutic approach for ccRCC.
Collapse
Affiliation(s)
- Luyao Chen
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yongpeng Xie
- Department of Urology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Ma
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Yu Zhang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Xintao Li
- Department of Urology, Chinese PLA Air Force General Hospital, Beijing, China
| | - Fan Zhang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Yu Gao
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Yang Fan
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Liangyou Gu
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Lei Wang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Xu Zhang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Bin Fu
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
44
|
Zhang J, Huang YJ, Yoon JY, Kemmitt J, Wright C, Schneider K, Sphabmixay P, Hernandez-Gordillo V, Holcomb SJ, Bhushan B, Rohatgi G, Benton K, Carpenter D, Kester JC, Eng G, Breault DT, Yilmaz O, Taketani M, Voigt CA, Carrier RL, Trumper DL, Griffith LG. Primary human colonic mucosal barrier crosstalk with super oxygen-sensitive Faecalibacterium prausnitzii in continuous culture. MED 2020; 2:74-98.e9. [PMID: 33511375 DOI: 10.1016/j.medj.2020.07.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background The gut microbiome plays an important role in human health and disease. Gnotobiotic animal and in vitro cell-based models provide some informative insights into mechanistic crosstalk. However, there is no existing system for a long-term co-culture of a human colonic mucosal barrier with super oxygen-sensitive commensal microbes, hindering the study of human-microbe interactions in a controlled manner. Methods Here, we investigated the effects of an abundant super oxygen-sensitive commensal anaerobe, Faecalibacterium prausnitzii, on a primary human mucosal barrier using a Gut-MIcrobiome (GuMI) physiome platform that we designed and fabricated. Findings Long-term continuous co-culture of F. prausnitzii for two days with colon epithelia, enabled by continuous flow of completely anoxic apical media and aerobic basal media, resulted in a strictly anaerobic apical environment fostering growth of and butyrate production by F. prausnitzii, while maintaining a stable colon epithelial barrier. We identified elevated differentiation and hypoxia-responsive genes and pathways in the platform compared with conventional aerobic static culture of the colon epithelia, attributable to a combination of anaerobic environment and continuous medium replenishment. Furthermore, we demonstrated anti-inflammatory effects of F. prausnitzii through HDAC and the TLR-NFKB axis. Finally, we identified that butyrate largely contributes to the anti-inflammatory effects by downregulating TLR3 and TLR4. Conclusions Our results are consistent with some clinical observations regarding F. prausnitzii, thus motivating further studies employing this platform with more complex engineered colon tissues for understanding the interaction between the human colonic mucosal barrier and microbiota, pathogens, or engineered bacteria.
Collapse
Affiliation(s)
| | | | - Jun Young Yoon
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,School of Mechanical Engineering, Yonsei University, Seoul 03722, South Korea
| | | | | | | | | | | | | | - Brij Bhushan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gar Rohatgi
- EPAM Continuum, 41 University Drive, Newtown, PA 18940, USA
| | - Kyle Benton
- EPAM Continuum, 41 University Drive, Newtown, PA 18940, USA
| | | | | | | | - David T Breault
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Rebecca L Carrier
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - David L Trumper
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linda G Griffith
- Department of Biological Engineering.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
45
|
Foxh1/Nodal Defines Context-Specific Direct Maternal Wnt/β-Catenin Target Gene Regulation in Early Development. iScience 2020; 23:101314. [PMID: 32650116 PMCID: PMC7347983 DOI: 10.1016/j.isci.2020.101314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/20/2020] [Accepted: 06/20/2020] [Indexed: 12/19/2022] Open
Abstract
Although Wnt/β-catenin signaling is generally conserved and well understood, the regulatory mechanisms controlling context-specific direct Wnt target gene expression in development and disease are still unclear. The onset of zygotic gene transcription in early embryogenesis represents an ideal, accessible experimental system to investigate context-specific direct Wnt target gene regulation. We combine transcriptomics using RNA-seq with genome-wide β-catenin association using ChIP-seq to identify stage-specific direct Wnt target genes. We propose coherent feedforward regulation involving two distinct classes of direct maternal Wnt target genes, which differ both in expression and persistence of β-catenin association. We discover that genomic β-catenin association overlaps with Foxh1-associated regulatory sequences and demonstrate that direct maternal Wnt target gene expression requires Foxh1 function and Nodal/Tgfβ signaling. Our results support a new paradigm for direct Wnt target gene co-regulation with context-specific mechanisms that will inform future studies of embryonic development and more widely stem cell-mediated homeostasis and human disease. Combining RNA-seq and β-catenin ChIP-seq identifies direct Wnt target genes Two distinct classes of direct maternal Wnt/β-catenin target genes can be discerned We propose coherent feedforward regulation of gene expression of the second class Maternal Wnt target gene expression of both classes requires Nodal/Foxh1 signaling
Collapse
|
46
|
Alahdal M, Duan L, Ouyang H, Wang D. The role of indoleamine 2,3 dioxygenase 1 in the osteoarthritis. Am J Transl Res 2020; 12:2322-2343. [PMID: 32655775 PMCID: PMC7344072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/05/2020] [Indexed: 06/11/2023]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease and a leading cause of disability. It involves articular cartilage destruction and a whole joint inflammation. In spite of OA pathogenesis is still unclear, new studies on the OA pathophysiological aetiology and immunomodulation therapy continuously achieve significant advances with new concepts. Here, we focus on the indoleamine-2,3-dioxygenase1 (IDO1) activity in the osteoarthritis (OA), which is one of the noticeable enzymes in the synovial fluid of arthritis patients. It was recognized as an essential mediator of autoreactive B and T cell responses in rheumatoid arthritis (RA) and an interesting therapeutic target against RA. However, the role IDO1 plays in the OA pathogenesis hasn't been discussed. The new OA experimental analysis evidenced IDO1 overexpression in the synovial fluid of OA patients, and recent studies reported that IDO1 metabolites were found higher in the OA synovial fluid than RA and spondyloarthropathies (SpA) patients. Moreover, the positive relation of IDO1 metabolites with OA pain and joint stiffness has been confirmed. Thus, the IDO1 plays a pivotal role in the pathogenesis of OA. In this review, the role IDO1 plays in the OA pathogenesis has been deeply discussed. It could be a promising target in the immunotherapy of OA disease.
Collapse
Affiliation(s)
- Murad Alahdal
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen 518035, P. R. China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of MedicineHangzhou, P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen 518035, P. R. China
| | - Li Duan
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen 518035, P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen 518035, P. R. China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of MedicineHangzhou, P. R. China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen 518035, P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen 518035, P. R. China
| |
Collapse
|
47
|
Altered Transcription Factor Binding and Gene Bivalency in Islets of Intrauterine Growth Retarded Rats. Cells 2020; 9:cells9061435. [PMID: 32527043 PMCID: PMC7348746 DOI: 10.3390/cells9061435] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/30/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
Intrauterine growth retardation (IUGR), which induces epigenetic modifications and permanent changes in gene expression, has been associated with the development of type 2 diabetes. Using a rat model of IUGR, we performed ChIP-Seq to identify and map genome-wide histone modifications and gene dysregulation in islets from 2- and 10-week rats. IUGR induced significant changes in the enrichment of H3K4me3, H3K27me3, and H3K27Ac marks in both 2-wk and 10-wk islets, which were correlated with expression changes of multiple genes critical for islet function in IUGR islets. ChIP-Seq analysis showed that IUGR-induced histone mark changes were enriched at critical transcription factor binding motifs, such as C/EBPs, Ets1, Bcl6, Thrb, Ebf1, Sox9, and Mitf. These transcription factors were also identified as top upstream regulators in our previously published transcriptome study. In addition, our ChIP-seq data revealed more than 1000 potential bivalent genes as identified by enrichment of both H3K4me3 and H3K27me3. The poised state of many potential bivalent genes was altered by IUGR, particularly Acod1, Fgf21, Serpina11, Cdh16, Lrrc27, and Lrrc66, key islet genes. Collectively, our findings suggest alterations of histone modification in key transcription factors and genes that may contribute to long-term gene dysregulation and an abnormal islet phenotype in IUGR rats.
Collapse
|
48
|
Lin C, Ding J, Bar-Joseph Z. Inferring TF activation order in time series scRNA-Seq studies. PLoS Comput Biol 2020; 16:e1007644. [PMID: 32069291 PMCID: PMC7048296 DOI: 10.1371/journal.pcbi.1007644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 02/28/2020] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Methods for the analysis of time series single cell expression data (scRNA-Seq) either do not utilize information about transcription factors (TFs) and their targets or only study these as a post-processing step. Using such information can both, improve the accuracy of the reconstructed model and cell assignments, while at the same time provide information on how and when the process is regulated. We developed the Continuous-State Hidden Markov Models TF (CSHMM-TF) method which integrates probabilistic modeling of scRNA-Seq data with the ability to assign TFs to specific activation points in the model. TFs are assumed to influence the emission probabilities for cells assigned to later time points allowing us to identify not just the TFs controlling each path but also their order of activation. We tested CSHMM-TF on several mouse and human datasets. As we show, the method was able to identify known and novel TFs for all processes, assigned time of activation agrees with both expression information and prior knowledge and combinatorial predictions are supported by known interactions. We also show that CSHMM-TF improves upon prior methods that do not utilize TF-gene interaction. An important attribute of time series single cell RNA-Seq (scRNA-Seq) data, is the ability to infer continuous trajectories of genes based on orderings of the cells. While several methods have been developed for ordering cells and inferring such trajectories, to date it was not possible to use these to infer the temporal activity of several key TFs. These TFs are are only post-transcriptionally regulated and so their expression does not provide complete information on their activity. To address this we developed the Continuous-State Hidden Markov Models TF (CSHMM-TF) methods that assigns continuous activation time to TFs based on both, their expression and the expression of their targets. Applying our method to several time series scRNA-Seq datasets we show that it correctly identifies the key regulators for the processes being studied. We analyze the temporal assignments for these TFs and show that they provide new insights about combinatorial regulation and the ordering of TF activation. We used several complementary sources to validate some of these predictions and discuss a number of other novel suggestions based on the method. As we show, the method is able to scale to large and noisy datasets and so is appropriate for several studies utilizing time series scRNA-Seq data.
Collapse
Affiliation(s)
- Chieh Lin
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Jun Ding
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Ziv Bar-Joseph
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
49
|
Kim E, Jung S, Wu Z, Zhang S, Jung H. Sox2 maintains epithelial cell proliferation in the successional dental lamina. Cell Prolif 2020; 53:e12729. [PMID: 31746095 PMCID: PMC6985665 DOI: 10.1111/cpr.12729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/10/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES The successional dental lamina is the distinctive structure on the lingual side of the vertebrate tooth germ. The aim of this study was to investigate the relationship among Sox2, Claudin10 and laminin5 and the role of Sox2 in successional dental lamina proliferation during vertebrate tooth development. MATERIALS AND METHODS To understand the successional dental lamina, two types of successional tooth formation, that in geckos (with multiple rounds of tooth generation) and that in mice (with only one round of tooth generation), were analysed. RESULTS Unique coexpression patterns of Sox2 and Claudin10 expression were compared in the successional dental lamina from the cap stage to the late bell stage in the mouse tooth germ and in juvenile gecko teeth to support continuous tooth replacement. Furthermore, Laminin5 expression was shown in the cap stage and decreased after the bell stage. Upon comparing the epithelial cell cycles and cell proliferation in successional dental lamina regions between mouse and gecko molars using BrdU and IdU staining and pulse-chase methods, distinctive patterns of continuous expression were revealed. Moreover, Sox2 overexpression with a lentiviral system resulted in hyperplastic dental epithelium in mouse molars. CONCLUSIONS Our findings indicate that the regulation of Sox2 in dental lamina proliferation is fundamental to the successional dental lamina in both species.
Collapse
Affiliation(s)
- Eun‐Jung Kim
- Division in Anatomy and Developmental BiologyDepartment of Oral BiologyResearch Center for Orofacial Hard Tissue RegenerationBrain Korea 21 PLUS ProjectOral Science Research CenterCollege of DentistryYonsei UniversitySeoulKorea
| | - Seo‐Yoon Jung
- Division in Anatomy and Developmental BiologyDepartment of Oral BiologyResearch Center for Orofacial Hard Tissue RegenerationBrain Korea 21 PLUS ProjectOral Science Research CenterCollege of DentistryYonsei UniversitySeoulKorea
| | - Zhaoming Wu
- Division in Anatomy and Developmental BiologyDepartment of Oral BiologyResearch Center for Orofacial Hard Tissue RegenerationBrain Korea 21 PLUS ProjectOral Science Research CenterCollege of DentistryYonsei UniversitySeoulKorea
| | - Sushan Zhang
- Division in Anatomy and Developmental BiologyDepartment of Oral BiologyResearch Center for Orofacial Hard Tissue RegenerationBrain Korea 21 PLUS ProjectOral Science Research CenterCollege of DentistryYonsei UniversitySeoulKorea
| | - Han‐Sung Jung
- Division in Anatomy and Developmental BiologyDepartment of Oral BiologyResearch Center for Orofacial Hard Tissue RegenerationBrain Korea 21 PLUS ProjectOral Science Research CenterCollege of DentistryYonsei UniversitySeoulKorea
| |
Collapse
|
50
|
Corada M, Orsenigo F, Bhat GP, Conze LL, Breviario F, Cunha SI, Claesson-Welsh L, Beznoussenko GV, Mironov AA, Bacigaluppi M, Martino G, Pitulescu ME, Adams RH, Magnusson P, Dejana E. Fine-Tuning of Sox17 and Canonical Wnt Coordinates the Permeability Properties of the Blood-Brain Barrier. Circ Res 2019; 124:511-525. [PMID: 30591003 PMCID: PMC6407809 DOI: 10.1161/circresaha.118.313316] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Supplemental Digital Content is available in the text. Rationale: The microvasculature of the central nervous system includes the blood-brain barrier (BBB), which regulates the permeability to nutrients and restricts the passage of toxic agents and inflammatory cells. Canonical Wnt/β-catenin signaling is responsible for the early phases of brain vascularization and BBB differentiation. However, this signal declines after birth, and other signaling pathways able to maintain barrier integrity at postnatal stage are still unknown. Objective: Sox17 (SRY [sex-determining region Y]-box 17) constitutes a major downstream target of Wnt/β-catenin in endothelial cells and regulates arterial differentiation. In the present article, we asked whether Sox17 may act downstream of Wnt/β-catenin in inducing BBB differentiation and maintenance. Methods and Results: Using reporter mice and nuclear staining of Sox17 and β-catenin, we report that although β-catenin signaling declines after birth, Sox17 activation increases and remains high in the adult. Endothelial-specific inactivation of Sox17 leads to increase of permeability of the brain microcirculation. The severity of this effect depends on the degree of BBB maturation: it is strong in the embryo and progressively declines after birth. In search of Sox17 mechanism of action, RNA sequencing analysis of gene expression of brain endothelial cells has identified members of the Wnt/β-catenin signaling pathway as downstream targets of Sox17. Consistently, we found that Sox17 is a positive inducer of Wnt/β-catenin signaling, and it acts in concert with this pathway to induce and maintain BBB properties. In vivo, inhibition of the β-catenin destruction complex or expression of a degradation-resistant β-catenin mutant, prevent the increase in permeability and retina vascular malformations observed in the absence of Sox17. Conclusions: Our data highlight a novel role for Sox17 in the induction and maintenance of the BBB, and they underline the strict reciprocal tuning of this transcription factor and Wnt/β-catenin pathway. Modulation of Sox17 activity may be relevant to control BBB permeability in pathological conditions.
Collapse
Affiliation(s)
- Monica Corada
- From the FIRC Institute of Molecular Oncology Foundation (IFOM), Milan, Italy (M.C., F.O., F.B., G.V.B., A.A.M., E.D.)
| | - Fabrizio Orsenigo
- From the FIRC Institute of Molecular Oncology Foundation (IFOM), Milan, Italy (M.C., F.O., F.B., G.V.B., A.A.M., E.D.)
| | - Ganesh Parameshwar Bhat
- Molecular Neurobiology Laboratory, Division of Neuroscience (G.P.B.), San Raffaele Hospital, Milan, Italy
| | - Lei Liu Conze
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (L.L.C., S.I.C., L.C.-W., P.M., E.D.)
| | - Ferruccio Breviario
- From the FIRC Institute of Molecular Oncology Foundation (IFOM), Milan, Italy (M.C., F.O., F.B., G.V.B., A.A.M., E.D.)
| | - Sara Isabel Cunha
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (L.L.C., S.I.C., L.C.-W., P.M., E.D.)
| | - Lena Claesson-Welsh
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (L.L.C., S.I.C., L.C.-W., P.M., E.D.)
| | - Galina V Beznoussenko
- From the FIRC Institute of Molecular Oncology Foundation (IFOM), Milan, Italy (M.C., F.O., F.B., G.V.B., A.A.M., E.D.)
| | - Alexander A Mironov
- From the FIRC Institute of Molecular Oncology Foundation (IFOM), Milan, Italy (M.C., F.O., F.B., G.V.B., A.A.M., E.D.)
| | - Marco Bacigaluppi
- Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology (M.B., G.M.), San Raffaele Hospital, Milan, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology (M.B., G.M.), San Raffaele Hospital, Milan, Italy
| | - Mara E Pitulescu
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and Faculty of Medicine, University of Münster, Germany (M.E.P., R.H.A.)
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and Faculty of Medicine, University of Münster, Germany (M.E.P., R.H.A.)
| | - Peetra Magnusson
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (L.L.C., S.I.C., L.C.-W., P.M., E.D.)
| | - Elisabetta Dejana
- From the FIRC Institute of Molecular Oncology Foundation (IFOM), Milan, Italy (M.C., F.O., F.B., G.V.B., A.A.M., E.D.).,Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (L.L.C., S.I.C., L.C.-W., P.M., E.D.).,Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| |
Collapse
|