1
|
Schoultz E, Moccia C, Liang S, Johansson E, Nilsson M. Tumor Cell Plasticity and Stromal Microenvironment Distinguish Papillary and Follicular Growth Patterns in a Mouse Model of BRAFV600E-Induced Thyroid Cancer. CANCER RESEARCH COMMUNICATIONS 2025; 5:409-421. [PMID: 39956582 PMCID: PMC11885905 DOI: 10.1158/2767-9764.crc-24-0474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/27/2024] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
SIGNIFICANCE Cell-of-origin intrinsic features rather than driver mutation identity influence tumor growth patterning in differentiated thyroid cancer and might impact histopathologic diagnosis of thyroid carcinoma subtypes.
Collapse
Affiliation(s)
- Elin Schoultz
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Carmen Moccia
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Shawn Liang
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Ellen Johansson
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Mikael Nilsson
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
2
|
Hrncir HR, Goodloe B, Bombin S, Hogan CB, Jadi O, Gracz AD. Sox9 inhibits Activin A to promote biliary maturation and branching morphogenesis. Nat Commun 2025; 16:1667. [PMID: 39955269 PMCID: PMC11830073 DOI: 10.1038/s41467-025-56813-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025] Open
Abstract
Intrahepatic bile duct (IHBD) development produces a morphologically heterogeneous network of large "ducts" and small "ductules" by adulthood. IHBD formation is closely linked to developmental specification of biliary epithelial cells (BECs) starting as early as E13.5, but mechanisms regulating differential IHBD morphology remain poorly understood. Here, we show that duct and ductule development has distinct genetic requirements, with Sox9 required to form the developmental precursors to peripheral ductules in adult livers. By optimizing large-volume IHBD imaging, we find that IHBDs emerge as a homogeneous webbed structure by E15.5 and undergo morphological maturation through 2 weeks of age. Developmental knockout of Sox9 leads to decreased postnatal branching morphogenesis, resulting in adult IHBDs with normal ducts but significantly fewer ductules. In the absence of Sox9, BECs fail to mature and exhibit elevated TGF-β signaling and Activin A. Exogenous Activin A is sufficient to induce developmental gene expression and morphological defects in wild-type BEC organoids, while early postnatal inhibition of Activin A in vivo rescues IHBD morphogenesis in the absence of Sox9. Our data demonstrate that proper IHBD architecture relies on inhibition of Activin A by Sox9 to promote ductule morphogenesis, defining regulatory mechanisms underlying morphological heterogeneity.
Collapse
Affiliation(s)
- Hannah R Hrncir
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA, USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA, USA
| | - Brianna Goodloe
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA, USA
| | - Sergei Bombin
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA, USA
| | - Connor B Hogan
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA, USA
| | - Othmane Jadi
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam D Gracz
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, GA, USA.
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
3
|
Fang Y, Wan JP, Wang Z, Song SY, Zhang CX, Yang L, Zhang QY, Yan CY, Wu FY, Lu SY, Sun F, Han B, Zhao SX, Dong M, Song HD. Deficiency of the HGF/Met pathway leads to thyroid dysgenesis by impeding late thyroid expansion. Nat Commun 2024; 15:3165. [PMID: 38605010 PMCID: PMC11009301 DOI: 10.1038/s41467-024-47363-9] [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: 06/17/2023] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
The mechanisms of bifurcation, a key step in thyroid development, are largely unknown. Here we find three zebrafish lines from a forward genetic screening with similar thyroid dysgenesis phenotypes and identify a stop-gain mutation in hgfa and two missense mutations in met by positional cloning from these zebrafish lines. The elongation of the thyroid primordium along the pharyngeal midline was dramatically disrupted in these zebrafish lines carrying a mutation in hgfa or met. Further studies show that MAPK inhibitor U0126 could mimic thyroid dysgenesis in zebrafish, and the phenotypes are rescued by overexpression of constitutively active MEK or Snail, downstream molecules of the HGF/Met pathway, in thyrocytes. Moreover, HGF promotes thyrocyte migration, which is probably mediated by downregulation of E-cadherin expression. The delayed bifurcation of the thyroid primordium is also observed in thyroid-specific Met knockout mice. Together, our findings reveal that HGF/Met is indispensable for the bifurcation of the thyroid primordium during thyroid development mediated by downregulation of E-cadherin in thyrocytes via MAPK-snail pathway.
Collapse
Affiliation(s)
- Ya Fang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, Jiangsu, 215000, China
| | - Jia-Ping Wan
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zheng Wang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shi-Yang Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cao-Xu Zhang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Liu Yang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qian-Yue Zhang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chen-Yan Yan
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Feng-Yao Wu
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Sang-Yu Lu
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Feng Sun
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bing Han
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shuang-Xia Zhao
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Mei Dong
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Huai-Dong Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
4
|
Hrncir HR, Bombin S, Goodloe B, Hogan CB, Jadi O, Gracz AD. Sox9 links biliary maturation to branching morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.574730. [PMID: 38293117 PMCID: PMC10827067 DOI: 10.1101/2024.01.15.574730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Branching morphogenesis couples cellular differentiation with development of tissue architecture. Intrahepatic bile duct (IHBD) morphogenesis is initiated with biliary epithelial cell (BEC) specification and eventually forms a heterogeneous network of large ducts and small ductules. Here, we show that Sox9 is required for developmental establishment of small ductules. IHBDs emerge as a webbed structure by E15.5 and undergo morphological maturation through 2 weeks of age. Developmental knockout of Sox9 leads to decreased postnatal branching morphogenesis, manifesting as loss of ductules in adult livers. In the absence of Sox9, BECs fail to mature and exhibit elevated TGF-β signaling and Activin A. Activin A induces developmental gene expression and morphological defects in BEC organoids and represses ductule formation in postnatal livers. Our data demonstrate that adult IHBD morphology and BEC maturation is regulated by the Sox9-dependent formation of precursors to ductules during development, mediated in part by downregulation of Activin A.
Collapse
Affiliation(s)
- Hannah R Hrncir
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University
| | - Sergei Bombin
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
| | - Brianna Goodloe
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
| | - Connor B Hogan
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
| | - Othmane Jadi
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Adam D Gracz
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University
- Lead contact:
| |
Collapse
|
5
|
Haneda Y, Miyagawa-Tomita S, Uchijima Y, Iwase A, Asai R, Kohro T, Wada Y, Kurihara H. Diverse contribution of amniogenic somatopleural cells to cardiovascular development: With special reference to thyroid vasculature. Dev Dyn 2024; 253:59-77. [PMID: 36038963 DOI: 10.1002/dvdy.532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The somatopleure serves as the primordium of the amnion, an extraembryonic membrane surrounding the embryo. Recently, we have reported that amniogenic somatopleural cells (ASCs) not only form the amnion but also migrate into the embryo and differentiate into cardiomyocytes and vascular endothelial cells. However, detailed differentiation processes and final distributions of these intra-embryonic ASCs (hereafter referred to as iASCs) remain largely unknown. RESULTS By quail-chick chimera analysis, we here show that iASCs differentiate into various cell types including cardiomyocytes, smooth muscle cells, cardiac interstitial cells, and vascular endothelial cells. In the pharyngeal region, they distribute selectively into the thyroid gland and differentiate into vascular endothelial cells to form intra-thyroid vasculature. Explant culture experiments indicated sequential requirement of fibroblast growth factor (FGF) and vascular endothelial growth factor (VEGF) signaling for endothelial differentiation of iASCs. Single-cell transcriptome analysis further revealed heterogeneity and the presence of hemangioblast-like cell population within ASCs, with a switch from FGF to VEGF receptor gene expression. CONCLUSION The present study demonstrates novel roles of ASCss especially in heart and thyroid development. It will provide a novel clue for understanding the cardiovascular development of amniotes from embryological and evolutionary perspectives.
Collapse
Affiliation(s)
- Yuka Haneda
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Sachiko Miyagawa-Tomita
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Animal Nursing Science, Yamazaki University of Animal Health Technology, Tokyo, Japan
| | - Yasunobu Uchijima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akiyasu Iwase
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Rieko Asai
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, USA
| | - Takahide Kohro
- Department of Medical Informatics, Jichi Medical University, Tochigi, Japan
| | - Youichiro Wada
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Yang RM, Song SY, Wu FY, Yang RF, Shen YT, Tu PH, Wang Z, Zhang JX, Cheng F, Gao GQ, Liang J, Guo MM, Yang L, Zhou Y, Zhao SX, Zhan M, Song HD. Myeloid cells interact with a subset of thyrocytes to promote their migration and follicle formation through NF-κB. Nat Commun 2023; 14:8082. [PMID: 38057310 PMCID: PMC10700497 DOI: 10.1038/s41467-023-43895-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/23/2023] [Indexed: 12/08/2023] Open
Abstract
The pathogenesis of thyroid dysgenesis (TD) is not well understood. Here, using a combination of single-cell RNA and spatial transcriptome sequencing, we identify a subgroup of NF-κB-activated thyrocytes located at the center of thyroid tissues in postnatal mice, which maintained a partially mesenchymal phenotype. These cells actively protruded out of the thyroid primordium and generated new follicles in zebrafish embryos through continuous tracing. Suppressing NF-κB signaling affected thyrocyte migration and follicle formation, leading to a TD-like phenotype in both mice and zebrafish. Interestingly, during thyroid folliculogenesis, myeloid cells played a crucial role in promoting thyrocyte migration by maintaining close contact and secreting TNF-α. We found that cebpa mutant zebrafish, in which all myeloid cells were depleted, exhibited thyrocyte migration defects. Taken together, our results suggest that myeloid-derived TNF-α-induced NF-κB activation plays a critical role in promoting the migration of vertebrate thyrocytes for follicle generation.
Collapse
Affiliation(s)
- Rui-Meng Yang
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shi-Yang Song
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng-Yao Wu
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Feng Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan-Ting Shen
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping-Hui Tu
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun-Xiu Zhang
- Department of Endocrinology, Maternal and Child Health Institute of Bozhou, Bozhou, China
| | - Feng Cheng
- Department of Laboratory Medicine, Fujian Children's Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Guan-Qi Gao
- Department of Endocrinology, The Linyi People's Hospital, Linyi, Shandong Province, China
| | - Jun Liang
- Department of Endocrinology, The Central Hospital of Xuzhou Affiliated to Xuzhou Medical College, Xuzhou, China
| | - Miao-Miao Guo
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liu Yang
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhou
- Stem Cell Program, Boston Children's Hospital and Harvard Stem Cell Institute, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | - Shuang-Xia Zhao
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ming Zhan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Huai-Dong Song
- Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Ryser MD, Greenwald MA, Sorribes IC, King LM, Hall A, Geradts J, Weaver DL, Mallo D, Holloway S, Monyak D, Gumbert G, Vaez-Ghaemi S, Wu E, Murgas K, Grimm LJ, Maley CC, Marks JR, Shibata D, Hwang ES. Growth Dynamics of Ductal Carcinoma in Situ Recapitulate Normal Breast Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.01.560370. [PMID: 37873488 PMCID: PMC10592867 DOI: 10.1101/2023.10.01.560370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Ductal carcinoma in situ (DCIS) and invasive breast cancer share many morphologic, proteomic, and genomic alterations. Yet in contrast to invasive cancer, many DCIS tumors do not progress and may remain indolent over decades. To better understand the heterogenous nature of this disease, we reconstructed the growth dynamics of 18 DCIS tumors based on the geo-spatial distribution of their somatic mutations. The somatic mutation topographies revealed that DCIS is multiclonal and consists of spatially discontinuous subclonal lesions. Here we show that this pattern of spread is consistent with a new 'Comet' model of DCIS tumorigenesis, whereby multiple subclones arise early and nucleate the buds of the growing tumor. The discontinuous, multiclonal growth of the Comet model is analogous to the branching morphogenesis of normal breast development that governs the rapid expansion of the mammary epithelium during puberty. The branching morphogenesis-like dynamics of the proposed Comet model diverges from the canonical model of clonal evolution, and better explains observed genomic spatial data. Importantly, the Comet model allows for the clinically relevant scenario of extensive DCIS spread, without being subjected to the selective pressures of subclone competition that promote the emergence of increasingly invasive phenotypes. As such, the normal cell movement inferred during DCIS growth provides a new explanation for the limited risk of progression in DCIS and adds biologic rationale for ongoing clinical efforts to reduce DCIS overtreatment.
Collapse
Affiliation(s)
- Marc D. Ryser
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
- Department of Mathematics, Duke University, Durham, NC, USA
| | | | | | - Lorraine M. King
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Allison Hall
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Joseph Geradts
- Department of Pathology, East Carolina University School of Medicine, Greenville, NC, USA
| | - Donald L. Weaver
- Larner College of Medicine, University of Vermont and UVM Cancer Center, Burlington, VT, USA
| | - Diego Mallo
- Arizona Cancer Evolution Center, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Shannon Holloway
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Daniel Monyak
- Trinity College of Arts and Sciences, Duke University, Durham, NC
| | - Graham Gumbert
- Trinity College of Arts and Sciences, Duke University, Durham, NC
| | | | - Ethan Wu
- Trinity College of Arts and Sciences, Duke University, Durham, NC
| | - Kevin Murgas
- Department of Biomedical Informatics, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Lars J. Grimm
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Carlo C. Maley
- Arizona Cancer Evolution Center, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Jeffrey R. Marks
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Darryl Shibata
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - E. Shelley Hwang
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
8
|
Schoultz E, Liang S, Carlsson T, Filges S, Ståhlberg A, Fagman H, Wiel C, Sayin V, Nilsson M. Tissue specificity of oncogenic BRAF targeted to lung and thyroid through a shared lineage factor. iScience 2023; 26:107071. [PMID: 37534159 PMCID: PMC10391731 DOI: 10.1016/j.isci.2023.107071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 04/05/2023] [Accepted: 06/05/2023] [Indexed: 08/04/2023] Open
Abstract
Cells of origin in cancer determine tumor phenotypes, but whether lineage-defining transcription factors might influence tissue specificity of tumorigenesis among organs with similar developmental traits are unknown. We demonstrate here that tumor development and progression markedly differ in lung and thyroid targeted by Braf mutation in Nkx2.1CreERT2 mice heterozygous for Nkx2-1. In absence of tamoxifen, non-induced Nkx2.1CreERT2;BrafCA/+ mutants developed multiple full-blown lung adenocarcinomas with a latency of 1-3 months whereas thyroid tumors were rare and constrained, although minute BrafCA activation documented by variant allele sequencing was similar in both tissues. Induced oncogene activation accelerated neoplastic growth only in the lungs. By contrast, NKX2-1+ progenitor cells were equally responsive to constitutive expression of mutant Braf during lung and thyroid development. Both lung and thyroid cells transiently downregulated NKX2-1 in early tumor stages. These results indicate that BRAFV600E-induced tumorigenesis obey organ-specific traits that might be differentially modified by a shared lineage factor.
Collapse
Affiliation(s)
- Elin Schoultz
- Sahlgrenska Center for Cancer Research, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Shawn Liang
- Sahlgrenska Center for Cancer Research, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Therese Carlsson
- Sahlgrenska Center for Cancer Research, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Stefan Filges
- Sahlgrenska Center for Cancer Research, University of Gothenburg, Göteborg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, University of Gothenburg, Göteborg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Göteborg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Göteborg, Sweden
| | - Henrik Fagman
- Sahlgrenska Center for Cancer Research, University of Gothenburg, Göteborg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Clotilde Wiel
- Sahlgrenska Center for Cancer Research, University of Gothenburg, Göteborg, Sweden
- Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, Göteborg, Sweden
| | - Volkan Sayin
- Sahlgrenska Center for Cancer Research, University of Gothenburg, Göteborg, Sweden
- Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, Göteborg, Sweden
| | - Mikael Nilsson
- Sahlgrenska Center for Cancer Research, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
9
|
Sox9 is involved in the thyroid differentiation program and is regulated by crosstalk between TSH, TGFβ and thyroid transcription factors. Sci Rep 2022; 12:2144. [PMID: 35140269 PMCID: PMC8828901 DOI: 10.1038/s41598-022-06004-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/21/2022] [Indexed: 11/09/2022] Open
Abstract
While the signaling pathways and transcription factors involved in the differentiation of thyroid follicular cells, both in embryonic and adult life, are increasingly well understood, the underlying mechanisms and potential crosstalk between the thyroid transcription factors Nkx2.1, Foxe1 and Pax8 and inductive signals remain unclear. Here, we focused on the transcription factor Sox9, which is expressed in Nkx2.1-positive embryonic thyroid precursor cells and is maintained from embryonic development to adulthood, but its function and control are unknown. We show that two of the main signals regulating thyroid differentiation, TSH and TGFβ, modulate Sox9 expression. Specifically, TSH stimulates the cAMP/PKA pathway to transcriptionally upregulate Sox9 mRNA and protein expression, a mechanism that is mediated by the binding of CREB to a CRE site within the Sox9 promoter. Contrastingly, TGFβ signals through Smad proteins to inhibit TSH-induced Sox9 transcription. Our data also reveal that Sox9 transcription is regulated by the thyroid transcription factors, particularly Pax8. Interestingly, Sox9 significantly increased the transcriptional activation of Pax8 and Foxe1 promoters and, consequently, their expression, but had no effect on Nkx2.1. Our study establishes the involvement of Sox9 in thyroid follicular cell differentiation and broadens our understanding of transcription factor regulation of thyroid function.
Collapse
|
10
|
Schoultz E, Johansson E, Moccia C, Jakubikova I, Ravi N, Liang S, Carlsson T, Montelius M, Patyra K, Kero J, Paulsson K, Fagman H, Bergo MO, Nilsson M. Tissue architecture delineates field cancerization in BRAFV600E-induced tumor development. Dis Model Mech 2022; 15:dmm048887. [PMID: 34379110 PMCID: PMC8380047 DOI: 10.1242/dmm.048887] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/24/2021] [Indexed: 12/29/2022] Open
Abstract
Cancer cells hijack developmental growth mechanisms but whether tissue morphogenesis and architecture modify tumorigenesis is unknown. Here, we characterized a new mouse model of sporadic thyroid carcinogenesis based on inducible expression of BRAF carrying a Val600 Glu (V600E) point mutation (BRAFV600E) from the thyroglobulin promoter (TgCreERT2). Spontaneous activation of this Braf-mutant allele due to leaky activity of the Cre recombinase revealed that intrinsic properties of thyroid follicles determined BRAF-mutant cell fate. Papillary thyroid carcinomas developed multicentrically within a normal microenvironment. Each tumor originated from a single follicle that provided a confined space for growth of a distinct tumor phenotype. Lineage tracing revealed oligoclonal tumor development in infancy and early selection of BRAFV600E kinase inhibitor-resistant clones. Somatic mutations were few, non-recurrent and limited to advanced tumors. Female mice developed larger tumors than males, reproducing the gender difference of human thyroid cancer. These data indicate that BRAFV600E-induced tumorigenesis is spatiotemporally regulated depending on the maturity and heterogeneity of follicles. Moreover, thyroid tissue organization seems to determine whether a BRAF-mutant lineage becomes a cancerized lineage. The TgCreERT2;BrafCA/+ sporadic thyroid cancer mouse model provides a new tool to evaluate drug therapy at different stages of tumor evolution.
Collapse
Affiliation(s)
- Elin Schoultz
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Ellen Johansson
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Carmen Moccia
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Iva Jakubikova
- Faculty of Medicine, Charles University, Hradec Kralove, Czech Republic
| | - Naveen Ravi
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund SE-22184, Sweden
| | - Shawn Liang
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Therese Carlsson
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| | - Mikael Montelius
- Department of Radiology, Institute of Clinical Sciences, University of Gothenburg, SE-41345 Göteborg, Sweden
| | - Konrad Patyra
- Department of Endocrinology, University of Turku, Åbo FI-20521, Finland
| | - Jukka Kero
- Department of Endocrinology, University of Turku, Åbo FI-20521, Finland
| | - Kajsa Paulsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund SE-22184, Sweden
| | - Henrik Fagman
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
- Department of Clinical Pathology, Sahlgrenska University Hospital, Göteborg SE-41345, Sweden
| | - Martin O. Bergo
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge SE-14183, Sweden
| | - Mikael Nilsson
- Sahlgrenska Center for Cancer Research, Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Göteborg, Sweden
| |
Collapse
|
11
|
Pu W, Shi X, Yu P, Zhang M, Liu Z, Tan L, Han P, Wang Y, Ji D, Gan H, Wei W, Lu Z, Qu N, Hu J, Hu X, Luo Z, Li H, Ji Q, Wang J, Zhang X, Wang YL. Single-cell transcriptomic analysis of the tumor ecosystems underlying initiation and progression of papillary thyroid carcinoma. Nat Commun 2021; 12:6058. [PMID: 34663816 PMCID: PMC8523550 DOI: 10.1038/s41467-021-26343-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 09/30/2021] [Indexed: 01/08/2023] Open
Abstract
The tumor ecosystem of papillary thyroid carcinoma (PTC) is poorly characterized. Using single-cell RNA sequencing, we profile transcriptomes of 158,577 cells from 11 patients’ paratumors, localized/advanced tumors, initially-treated/recurrent lymph nodes and radioactive iodine (RAI)-refractory distant metastases, covering comprehensive clinical courses of PTC. Our data identifies a “cancer-primed” premalignant thyrocyte population with normal morphology but altered transcriptomes. Along the developmental trajectory, we also discover three phenotypes of malignant thyrocytes (follicular-like, partial-epithelial-mesenchymal-transition-like, dedifferentiation-like), whose composition shapes bulk molecular subtypes, tumor characteristics and RAI responses. Furthermore, we uncover a distinct BRAF-like-B subtype with predominant dedifferentiation-like thyrocytes, enriched cancer-associated fibroblasts, worse prognosis and promising prospect of immunotherapy. Moreover, potential vascular-immune crosstalk in PTC provides theoretical basis for combined anti-angiogenic and immunotherapy. Together, our findings provide insight into the PTC ecosystem that suggests potential prognostic and therapeutic implications. The characterisation of the papillary thyroid carcinoma (PTC) tumour microenvironment remains crucial. Here, the authors perform single-cell RNA sequencing in 11 patients and identify potential opportunities for the use of immunotherapy and its combination with anti-angiogenic therapy in PTC.
Collapse
Affiliation(s)
- Weilin Pu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Xiao Shi
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Pengcheng Yu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Meiying Zhang
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhiyan Liu
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Licheng Tan
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peizhen Han
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Dongmei Ji
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Phase I Clinical Trial Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Hualei Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Wenjun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhongwu Lu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ning Qu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jiaqian Hu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaohua Hu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Zaili Luo
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Huajun Li
- Department of Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, 201210, China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438, China.,Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Xiaoming Zhang
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yu-Long Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Dom G, Dmitriev P, Lambot MA, Van Vliet G, Glinoer D, Libert F, Lefort A, Dumont JE, Maenhaut C. Transcriptomic Signature of Human Embryonic Thyroid Reveals Transition From Differentiation to Functional Maturation. Front Cell Dev Biol 2021; 9:669354. [PMID: 34249923 PMCID: PMC8270686 DOI: 10.3389/fcell.2021.669354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/17/2021] [Indexed: 11/16/2022] Open
Abstract
The human thyroid gland acquires a differentiation program as early as weeks 3-4 of embryonic development. The onset of functional differentiation, which manifests by the appearance of colloid in thyroid follicles, takes place during gestation weeks 10-11. By 12-13 weeks functional differentiation is accomplished and the thyroid is capable of producing thyroid hormones although at a low level. During maturation, thyroid hormones yield increases and physiological mechanisms of thyroid hormone synthesis regulation are established. In the present work we traced the process of thyroid functional differentiation and maturation in the course of human development by performing transcriptomic analysis of human thyroids covering the period of gestation weeks 7-11 and comparing it to adult human thyroid. We obtained specific transcriptomic signatures of embryonic and adult human thyroids by comparing them to non-thyroid tissues from human embryos and adults. We defined a non-TSH (thyroid stimulating hormone) dependent transition from differentiation to maturation of thyroid. The study also sought to shed light on possible factors that could replace TSH, which is absent in this window of gestational age, to trigger transition to the emergence of thyroid function. We propose a list of possible genes that may also be involved in abnormalities in thyroid differentiation and/or maturation, hence leading to congenital hypothyroidism. To our knowledge, this study represent the first transcriptomic analysis of human embryonic thyroid and its comparison to adult thyroid.
Collapse
Affiliation(s)
- Geneviève Dom
- School of Medicine, IRIBHM, Université libre de Bruxelles, Brussels, Belgium
- Institute of Interdisciplinary Research in Human and Molecular Biology, Brussels, Belgium
| | - Petr Dmitriev
- School of Medicine, IRIBHM, Université libre de Bruxelles, Brussels, Belgium
- Institute of Interdisciplinary Research in Human and Molecular Biology, Brussels, Belgium
| | | | - Guy Van Vliet
- Département de Pédiatrie, Université de Montréal, Montreal, QC, Canada
- CHU Sainte-Justine, Montreal, QC, Canada
| | - Daniel Glinoer
- Hôpital Saint-Pierre, Université libre de Bruxelles, Brussels, Belgium
| | | | - Anne Lefort
- School of Medicine, IRIBHM, Université libre de Bruxelles, Brussels, Belgium
| | - Jacques E. Dumont
- School of Medicine, IRIBHM, Université libre de Bruxelles, Brussels, Belgium
- Institute of Interdisciplinary Research in Human and Molecular Biology, Brussels, Belgium
| | - Carine Maenhaut
- School of Medicine, IRIBHM, Université libre de Bruxelles, Brussels, Belgium
- Institute of Interdisciplinary Research in Human and Molecular Biology, Brussels, Belgium
| |
Collapse
|
13
|
Johansson E, Liang S, Moccia C, Carlsson T, Andersson D, Fagman H, Nilsson M. Asynchrony of Apical Polarization, Luminogenesis, and Functional Differentiation in the Developing Thyroid Gland. Front Endocrinol (Lausanne) 2021; 12:760541. [PMID: 34975747 PMCID: PMC8719337 DOI: 10.3389/fendo.2021.760541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/12/2021] [Indexed: 12/02/2022] Open
Abstract
Follicular thyroid tissue originates from progenitors derived from a midline endodermal primordium. Current understanding infers that folliculogenesis in the embryonic thyroid designates the latest morphogenetic event taking place after the final anatomical shape and position of the gland is established. However, this concept does not consider the fact that the thyroid isthmus develops chronologically before the lobes and also contains all progenitors required for lobulation. To elucidate whether cells committed to a thyroid fate might be triggered to differentiate asynchronously related to maturation and developmental stage, mouse embryonic thyroid tissues from E12.5-17.5 were subjected to immunofluorescent labeling of biomarkers (progenitors: NKX2-1; differentiation: thyroglobulin/TG); folliculogenesis: E-cadherin/CDH1; luminogenesis: mucin 1/MUC1; apical polarity: pericentrin/PCNT; basement membrane: laminin; growth: Ki67), quantitative RT-PCR analysis (Nkx2.1, Tg, Muc1) and transmission electron microscopy. Tg expression was detectable as early as E12.5 and gradually increased >1000-fold until E17.5. Muc1 and Nkx2.1 transcript levels increased in the same time interval. Prior to lobulation (E12.5-13.5), MUC1 and TG distinguished pre-follicular from progenitor cells in the developing isthmus characterized by intense cell proliferation. Luminogenesis comprised redistribution of MUC1+ vesicles or vacuoles, transiently associated with PCNT, to the apical cytoplasm and the subsequent formation of MUC1+ nascent lumens. Apical polarization of pre-follicular cells and lumen initiation involved submembraneous vesicular traffic, reorganization of adherens junctions and ciliogenesis. MUC1 did not co-localize with TG until a lumen with a MUC1+ apical membrane was established. MUC1 delineated the lumen of all newly formed follicles encountered in the developing lobes at E15.5-17.5. Folliculogenesis started before establishment of a complete follicular basal lamina. These observations indicate that embryonic thyroid differentiation is an asynchronous process consistent with the idea that progenitors attaining a stationary position in the connecting isthmus portion undergo apical polarization and generate follicles already at a primordial stage of thyroid development, i.e. foregoing growth of the lobes. Although the thyroid isthmus eventually comprises minute amounts of the total thyroid volume and contributes little to the overall hormone production, it is of principal interest that local cues related to the residence status of cells - independently of a prevailing high multiplication rate - govern the thyroid differentiation program.
Collapse
Affiliation(s)
- Ellen Johansson
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Shawn Liang
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Carmen Moccia
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Therese Carlsson
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Daniel Andersson
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Henrik Fagman
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Clinical Pathology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Mikael Nilsson
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- *Correspondence: Mikael Nilsson,
| |
Collapse
|
14
|
Gonay L, Spourquet C, Baudoin M, Lepers L, Lemoine P, Fletcher AG, Hanert E, Pierreux CE. Modelling of Epithelial Growth, Fission and Lumen Formation During Embryonic Thyroid Development: A Combination of Computational and Experimental Approaches. Front Endocrinol (Lausanne) 2021; 12:655862. [PMID: 34163435 PMCID: PMC8216395 DOI: 10.3389/fendo.2021.655862] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/22/2021] [Indexed: 12/18/2022] Open
Abstract
Organogenesis is the phase of embryonic development leading to the formation of fully functional organs. In the case of the thyroid, organogenesis starts from the endoderm and generates a multitude of closely packed independent spherical follicular units surrounded by a dense network of capillaries. Follicular organisation is unique and essential for thyroid function, i.e. thyroid hormone production. Previous in vivo studies showed that, besides their nutritive function, endothelial cells play a central role during thyroid gland morphogenesis. However, the precise mechanisms and biological parameters controlling the transformation of the multi-layered thyroid epithelial primordium into a multitude of single-layered follicles are mostly unknown. Animal studies used to improve understanding of organogenesis are costly and time-consuming, with recognised limitations. Here, we developed and used a 2-D vertex model of thyroid growth, angiogenesis and folliculogenesis, within the open-source Chaste framework. Our in silico model, based on in vivo images, correctly simulates the differential growth and proliferation of central and peripheral epithelial cells, as well as the morphogen-driven migration of endothelial cells, consistently with our experimental data. Our simulations further showed that reduced epithelial cell adhesion was critical to allow endothelial invasion and fission of the multi-layered epithelial mass. Finally, our model also allowed epithelial cell polarisation and follicular lumen formation by endothelial cell abundance and proximity. Our study illustrates how constant discussion between theoretical and experimental approaches can help us to better understand the roles of cellular movement, adhesion and polarisation during thyroid embryonic development. We anticipate that the use of in silico models like the one we describe can push forward the fields of developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Leolo Gonay
- Earth and Life Institute, UCLouvain, Louvain-La-Neuve, Belgium
- de Duve Institute, UCLouvain, Woluwé-Saint-Lambert, Belgium
| | | | - Matthieu Baudoin
- Earth and Life Institute, UCLouvain, Louvain-La-Neuve, Belgium
- de Duve Institute, UCLouvain, Woluwé-Saint-Lambert, Belgium
| | - Ludovic Lepers
- Earth and Life Institute, UCLouvain, Louvain-La-Neuve, Belgium
- de Duve Institute, UCLouvain, Woluwé-Saint-Lambert, Belgium
| | | | - Alexander G. Fletcher
- School of Mathematics and Statistics, University of Sheffield, Sheffield, United Kingdom
| | - Emmanuel Hanert
- Earth and Life Institute, UCLouvain, Louvain-La-Neuve, Belgium
| | - Christophe E. Pierreux
- de Duve Institute, UCLouvain, Woluwé-Saint-Lambert, Belgium
- *Correspondence: Christophe E. Pierreux,
| |
Collapse
|
15
|
López-Márquez A, Carrasco-López C, Fernández-Méndez C, Santisteban P. Unraveling the Complex Interplay Between Transcription Factors and Signaling Molecules in Thyroid Differentiation and Function, From Embryos to Adults. Front Endocrinol (Lausanne) 2021; 12:654569. [PMID: 33959098 PMCID: PMC8095082 DOI: 10.3389/fendo.2021.654569] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/29/2021] [Indexed: 12/29/2022] Open
Abstract
Thyroid differentiation of progenitor cells occurs during embryonic development and in the adult thyroid gland, and the molecular bases of these complex and finely regulated processes are becoming ever more clear. In this Review, we describe the most recent advances in the study of transcription factors, signaling molecules and regulatory pathways controlling thyroid differentiation and development in the mammalian embryo. We also discuss the maintenance of the adult differentiated phenotype to ensure the biosynthesis of thyroid hormones. We will focus on endoderm-derived thyroid epithelial cells, which are responsible for the formation of the thyroid follicle, the functional unit of the thyroid gland. The use of animal models and pluripotent stem cells has greatly aided in providing clues to the complicated puzzle of thyroid development and function in adults. The so-called thyroid transcription factors - Nkx2-1, Foxe1, Pax8 and Hhex - were the first pieces of the puzzle identified in mice. Other transcription factors, either acting upstream of or directly with the thyroid transcription factors, were subsequently identified to, almost, complete the puzzle. Among them, the transcription factors Glis3, Sox9 and the cofactor of the Hippo pathway Taz, have emerged as important players in thyroid differentiation and development. The involvement of signaling molecules increases the complexity of the puzzle. In this context, the importance of Bmps, Fgfs and Shh signaling at the onset of development, and of TSH, IGF1 and TGFβ both at the end of terminal differentiation in embryos and in the adult thyroid, are well recognized. All of these aspects are covered herein. Thus, readers will be able to visualize the puzzle of thyroid differentiation with most - if not all - of the pieces in place.
Collapse
Affiliation(s)
- Arístides López-Márquez
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Laboratorio de Investigación Aplicada en Enfermedades Neuromusculares, Unidad de Patología Neuromuscular, Servicio de Neuropediatría, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Carlos Carrasco-López
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Celia Fernández-Méndez
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) y Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Pilar Santisteban,
| |
Collapse
|
16
|
Gillotay P, Shankar M, Haerlingen B, Sema Elif E, Pozo‐Morales M, Garteizgogeascoa I, Reinhardt S, Kränkel A, Bläsche J, Petzold A, Ninov N, Kesavan G, Lange C, Brand M, Lefort A, Libert F, Detours V, Costagliola S, Sumeet Pal S. Single-cell transcriptome analysis reveals thyrocyte diversity in the zebrafish thyroid gland. EMBO Rep 2020; 21:e50612. [PMID: 33140917 PMCID: PMC7726803 DOI: 10.15252/embr.202050612] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
The thyroid gland regulates growth and metabolism via production of thyroid hormone in follicles composed of thyrocytes. So far, thyrocytes have been assumed to be a homogenous population. To uncover heterogeneity in the thyrocyte population and molecularly characterize the non-thyrocyte cells surrounding the follicle, we developed a single-cell transcriptome atlas of the region containing the zebrafish thyroid gland. The 6249-cell atlas includes profiles of thyrocytes, blood vessels, lymphatic vessels, immune cells, and fibroblasts. Further, the thyrocytes show expression heterogeneity, including bimodal expression of the transcription factor pax2a. To validate thyrocyte heterogeneity, we generated a CRISPR/Cas9-based pax2a knock-in line that monitors pax2a expression in the thyrocytes. A population of pax2a-low mature thyrocytes interspersed in individual follicles can be distinguished. We corroborate heterogeneity within the thyrocyte population using RNA sequencing of pax2a-high and pax2a-low thyrocytes, which demonstrates 20% differential expression in transcriptome between the two subpopulations. Our results identify and validate transcriptional differences within the presumed homogenous thyrocyte population.
Collapse
Affiliation(s)
| | - Meghna Shankar
- IRIBHMUniversité Libre de Bruxelles (ULB)BrusselsBelgium
| | | | - Eski Sema Elif
- IRIBHMUniversité Libre de Bruxelles (ULB)BrusselsBelgium
| | | | | | - Susanne Reinhardt
- DRESDEN‐concept Genome CenterDFG NGS Competence Center, c/o Center for Molecular and Cellular BioengineeringTU DresdenDresdenGermany
| | - Annekathrin Kränkel
- DRESDEN‐concept Genome CenterDFG NGS Competence Center, c/o Center for Molecular and Cellular BioengineeringTU DresdenDresdenGermany
| | - Juliane Bläsche
- DRESDEN‐concept Genome CenterDFG NGS Competence Center, c/o Center for Molecular and Cellular BioengineeringTU DresdenDresdenGermany
| | - Andreas Petzold
- DRESDEN‐concept Genome CenterDFG NGS Competence Center, c/o Center for Molecular and Cellular BioengineeringTU DresdenDresdenGermany
| | - Nikolay Ninov
- Center for Regenerative Therapies Dresden (CRTD)TU DresdenDresdenGermany
| | - Gokul Kesavan
- Center for Regenerative Therapies Dresden TU Dresden (CRTD), and Cluster of ExcellencePhysics of Life (PoL)TU DresdenDresdenGermany
| | - Christian Lange
- Center for Regenerative Therapies Dresden TU Dresden (CRTD), and Cluster of ExcellencePhysics of Life (PoL)TU DresdenDresdenGermany
| | - Michael Brand
- Center for Regenerative Therapies Dresden TU Dresden (CRTD), and Cluster of ExcellencePhysics of Life (PoL)TU DresdenDresdenGermany
| | - Anne Lefort
- Center for Regenerative Therapies Dresden TU Dresden (CRTD), and Cluster of ExcellencePhysics of Life (PoL)TU DresdenDresdenGermany
| | - Frédérick Libert
- Center for Regenerative Therapies Dresden TU Dresden (CRTD), and Cluster of ExcellencePhysics of Life (PoL)TU DresdenDresdenGermany
| | | | | | | |
Collapse
|
17
|
Ran Q, Zhou Q, Oda K, Yasue A, Abe M, Ye X, Li Y, Sasaoka T, Sakimura K, Ajioka Y, Saijo Y. Generation of Thyroid Tissues From Embryonic Stem Cells via Blastocyst Complementation In Vivo. Front Endocrinol (Lausanne) 2020; 11:609697. [PMID: 33381086 PMCID: PMC7767966 DOI: 10.3389/fendo.2020.609697] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/10/2020] [Indexed: 01/29/2023] Open
Abstract
The generation of mature, functional, thyroid follicular cells from pluripotent stem cells would potentially provide a therapeutic benefit for patients with hypothyroidism, but in vitro differentiation remains difficult. We earlier reported the in vivo generation of lung organs via blastocyst complementation in fibroblast growth factor 10 (Fgf10), compound, heterozygous mutant (Fgf10 Ex1mut/Ex3mut) mice. Fgf10 also plays an essential role in thyroid development and branching morphogenesis, but any role thereof in thyroid organogenesis remains unclear. Here, we report that the thyroids of Fgf10 Ex1mut/Ex3mut mice exhibit severe hypoplasia, and we generate thyroid tissues from mouse embryonic stem cells (ESCs) in Fgf10 Ex1mut/Ex3mut mice via blastocyst complementation. The tissues were morphologically normal and physiologically functional. The thyroid follicular cells of Fgf10 Ex1mut/Ex3mut chimeric mice were derived largely from GFP-positive mouse ESCs although the recipient cells were mixed. Thyroid generation in vivo via blastocyst complementation will aid functional thyroid regeneration.
Collapse
Affiliation(s)
- Qingsong Ran
- Department of Medical Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Qiliang Zhou
- Department of Medical Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- *Correspondence: Qiliang Zhou,
| | - Kanako Oda
- Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akihiro Yasue
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Xulu Ye
- Department of Medical Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yingchun Li
- Department of Medical Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Toshikuni Sasaoka
- Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yoichi Ajioka
- Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yasuo Saijo
- Department of Medical Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
18
|
Haerlingen B, Opitz R, Vandernoot I, Trubiroha A, Gillotay P, Giusti N, Costagliola S. Small-Molecule Screening in Zebrafish Embryos Identifies Signaling Pathways Regulating Early Thyroid Development. Thyroid 2019; 29:1683-1703. [PMID: 31507237 DOI: 10.1089/thy.2019.0122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Defects in embryonic development of the thyroid gland are a major cause for congenital hypothyroidism in human newborns, but the underlying molecular mechanisms are still poorly understood. Organ development relies on a tightly regulated interplay between extrinsic signaling cues and cell intrinsic factors. At present, however, there is limited knowledge about the specific extrinsic signaling cues that regulate foregut endoderm patterning, thyroid cell specification, and subsequent morphogenetic processes in thyroid development. Methods: To begin to address this problem in a systematic way, we used zebrafish embryos to perform a series of in vivo phenotype-driven chemical genetic screens to identify signaling cues regulating early thyroid development. For this purpose, we treated zebrafish embryos during different developmental periods with a panel of small-molecule compounds known to manipulate the activity of major signaling pathways and scored phenotypic deviations in thyroid, endoderm, and cardiovascular development using whole-mount in situ hybridization and transgenic fluorescent reporter models. Results: Systematic assessment of drugged embryos recovered a range of thyroid phenotypes including expansion, reduction or lack of the early thyroid anlage, defective thyroid budding, as well as hypoplastic, enlarged, or overtly disorganized presentation of the thyroid primordium after budding. Our pharmacological screening identified bone morphogenetic protein and fibroblast growth factor signaling as key factors for thyroid specification and early thyroid organogenesis, highlighted the importance of low Wnt activities during early development for thyroid specification, and implicated drug-induced cardiac and vascular anomalies as likely indirect mechanisms causing various forms of thyroid dysgenesis. Conclusions: By integrating the outcome of our screening efforts with previously available information from other model organisms including Xenopus, chicken, and mouse, we conclude that signaling cues regulating thyroid development appear broadly conserved across vertebrates. We therefore expect that observations made in zebrafish can inform mammalian models of thyroid organogenesis to further our understanding of the molecular mechanisms of congenital thyroid diseases.
Collapse
Affiliation(s)
- Benoit Haerlingen
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Robert Opitz
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
- Institute of Experimental Pediatric Endocrinology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Isabelle Vandernoot
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Achim Trubiroha
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Pierre Gillotay
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Nicoletta Giusti
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Sabine Costagliola
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
19
|
Multiscale dynamics of branching morphogenesis. Curr Opin Cell Biol 2019; 60:99-105. [DOI: 10.1016/j.ceb.2019.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/09/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022]
|
20
|
Transcriptional control of lung alveolar type 1 cell development and maintenance by NK homeobox 2-1. Proc Natl Acad Sci U S A 2019; 116:20545-20555. [PMID: 31548395 DOI: 10.1073/pnas.1906663116] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The extraordinarily thin alveolar type 1 (AT1) cell constitutes nearly the entire gas exchange surface and allows passive diffusion of oxygen into the blood stream. Despite such an essential role, the transcriptional network controlling AT1 cells remains unclear. Using cell-specific knockout mouse models, genomic profiling, and 3D imaging, we found that NK homeobox 2-1 (Nkx2-1) is expressed in AT1 cells and is required for the development and maintenance of AT1 cells. Without Nkx2-1, developing AT1 cells lose 3 defining features-molecular markers, expansive morphology, and cellular quiescence-leading to alveolar simplification and lethality. NKX2-1 is also cell-autonomously required for the same 3 defining features in mature AT1 cells. Intriguingly, Nkx2-1 mutant AT1 cells activate gastrointestinal (GI) genes and form dense microvilli-like structures apically. Single-cell RNA-seq supports a linear transformation of Nkx2-1 mutant AT1 cells toward a GI fate. Whole lung ChIP-seq shows NKX2-1 binding to 68% of genes that are down-regulated upon Nkx2-1 deletion, including 93% of known AT1 genes, but near-background binding to up-regulated genes. Our results place NKX2-1 at the top of the AT1 cell transcriptional hierarchy and demonstrate remarkable plasticity of an otherwise terminally differentiated cell type.
Collapse
|
21
|
Adnan SH, Nobecourt P, Tran S, Radhakrishnan RS, Bowen-Jallow KA. Fourth branchial cleft cyst and congenital absence of the contralateral thyroid lobe. JOURNAL OF PEDIATRIC SURGERY CASE REPORTS 2019. [DOI: 10.1016/j.epsc.2019.101245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
22
|
Follicular cell lineage in persistent ultimobranchial remnants of mammals. Cell Tissue Res 2019; 376:1-18. [PMID: 30617614 DOI: 10.1007/s00441-018-02982-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023]
Abstract
It has been a subject of much debate whether thyroid follicular cells originate from the ultimobranchial body, in addition to median thyroid primordium. Ultimobranchial remnants are detected in normal dogs, rats, mice, cattle, bison and humans and also in mutant mice such as Eya1 homozygotes, Hox3 paralogs homozygotes, Nkx2.1 heterozygotes and FRS2α2F/2F. Besides C cells, follicular cell lineages immunoreactive for thyroglobulin are located within these ultimobranchial remnants. In dogs, the C cell complexes, i.e., large cell clusters consisting of C cells and undifferentiated cells, are present together with parathyroid IV and thymus IV in or close to the thyroid lobe. In addition, follicular cells in various stages of differentiation, including follicular cell groups and primitive and minute follicles storing colloid, are intermingled with C cells in some complexes. This review elaborates the transcription factors and signaling molecules involved in folliculogenesis and it is supposed why the follicular cells in the ultimobranchial remnants are sustained in immature stages. Pax8, a transcription factor crucial for the development of follicular cells, is expressed in the fourth pharyngeal pouch and the ultimobranchial body in human embryos. Pax8 expression is also detected in the ultimobranchial remnants of Eya1 and Hes1 null mutant mice. To determine whether the C cells and follicular cells in the ultimobranchial remnants consist of dual lineage cells or are derived from the common precursor, the changes of undifferentiated cells in dog C cell complexes are examined after chronically induced hypercalcemia or antithyroid drug treatment.
Collapse
|