1
|
Zhang Y, Ran L, Liu Y, Li W, Ran A, Li H, Huang B, Ren J, Ning H, Ma Q, Wang X, Yang F, Pan X, Liang C, Wang S, Qin C, Jiang Y, Qian K, Xiao B. FNDC3B promotes gastric cancer metastasis via interacting with FAM83H and preventing its proteasomal degradation. Cell Mol Biol Lett 2025; 30:65. [PMID: 40450207 DOI: 10.1186/s11658-025-00741-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/06/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most prevalent digestive tract malignancies, with metastasis being a major contributor to poor prognosis in patients. Fibronectin type III domain-containing 3B (FNDC3B) plays pivotal roles in various tumors, yet its role in GC remains unknown. METHODS We utilized a GC immunohistochemistry (IHC) chip and specimens to identify high expression of FNDC3B in GC. In vitro cellular experiments and in vivo nude mouse models were constructed to validate the biological functions of FNDC3B. Truncated mutants of FNDC3B and immunofluorescence (IF) assays were conducted to explore the critical domains of FNDC3B. Liquid chromatography-tandem mass spectrometry (LC-MS) and coimmunoprecipitation (Co-IP) were employed to screen and identify target proteins interacting with FNDC3B. Rescue experiments were performed to uncover interactions between FNDC3B and the binding protein. RESULTS FNDC3B was found to function as an oncogene in GC. Clinically, FNDC3B was significantly upregulated in GC specimens and associated with poor survival in patients with GC. Functionally, elevated FNDC3B promoted GC metastasis both in vitro and in vivo. Structurally, the proline-rich N-terminus and a transmembrane (TM) domain at the C-terminus were crucial for maintaining the metastasis function of FNDC3B. Mechanistically, FNDC3B interacted with FAM83H, inhibiting the ubiquitin-proteasome degradation of FAM83H, which in turn enhanced GC progression through the FNDC3B/FAM83H/Snail/EMT axis. CONCLUSIONS Our results demonstrate that FNDC3B promotes GC metastasis and has the potential to serve as a therapeutic target for GC.
Collapse
Affiliation(s)
- Yuying Zhang
- Department of Gastrointestinal Surgery of the First Affiliated Hospital and College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lingyu Ran
- Department of Kidney, Southwest Hospital, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Yuying Liu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wei Li
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Ai Ran
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Haiping Li
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Shaanxi Coal Chemical Technology Research Institute Co., Ltd, Xi'an, 710000, People's Republic of China
| | - Bo Huang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Junwu Ren
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hao Ning
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Qiang Ma
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaolin Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Feifei Yang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaojuan Pan
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Ce Liang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Shimin Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Changhong Qin
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yan Jiang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Kun Qian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Bin Xiao
- Department of Gastrointestinal Surgery of the First Affiliated Hospital and College of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
2
|
Kuga T, Saraya M, Higuchi S, Yoshida S, Murataka S, Fujiwara Y, Tomita Y, Hayama S, Kaibori Y, Yamagishi N. The DUF1669 domain of FAM83H is required for its localization to nuclear speckles. Sci Rep 2025; 15:12301. [PMID: 40210674 PMCID: PMC11986100 DOI: 10.1038/s41598-025-96356-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/27/2025] [Indexed: 04/12/2025] Open
Abstract
Autosomal-dominant hypocalcified amelogenesis imperfecta (ADHCAI) is caused by mutations in the FAM83H gene. Mutated FAM83H genes encode truncated FAM83H proteins with amino acid lengths between amino acids 1-286 and 1-693, in contrast to wild-type FAM83H (1-1179). Deletion of the C-terminus of FAM83H results in its subcellular translocation from the cytoplasmic compartment to the nuclear speckles, where splicing factors accumulate. However, the amino acid region of FAM83H required for nuclear speckle localization has not yet been determined, and whether all FAM83H-truncated proteins associated with ADHCAI localize to nuclear speckles remains unknown. Here, we examined the subcellular localization of FAM83H mutant proteins with truncations or deletions at various amino acid positions. Deletions within residues 1-300, which corresponds to the DUF1669 domain (17-281), attenuated or abolished the nuclear speckle localization of FAM83H. Meanwhile, some ADHCAI-related FAM83H-truncated proteins did not localize to nuclear speckles, despite the presence of the DUF1669 domain. These results suggest that the DUF1669 domain is required, but not sufficient, for nuclear speckle localization of FAM83H, demonstrating that nuclear speckle localization is not a common feature among FAM83H-truncated proteins related to ADHCAI.
Collapse
Affiliation(s)
- Takahisa Kuga
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan.
| | - Minami Saraya
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan
| | - Sora Higuchi
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan
| | - Shun Yoshida
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan
| | - Shino Murataka
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan
| | - Yuri Fujiwara
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan
| | - Yudai Tomita
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan
| | - Sayo Hayama
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan
| | - Yuichiro Kaibori
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan
| | - Nobuyuki Yamagishi
- Laboratory of Analytics for Biomolecules, Faculty of Pharmaceutical Science, Setsunan University, 45-1 Nagaotoge-cho, Hirakata-shi, 573-0101, Osaka, Japan
| |
Collapse
|
3
|
Tynior W, Hudy D, Gołąbek K, Raczkowska-Siostrzonek A, Strzelczyk JK. Expression of AMELX, AMBN, ENAM, TUFT1, FAM83H and MMP20 Genes in Buccal Epithelial Cells from Patients with Molar Incisor Hypomineralization (MIH)-A Pilot Study. Int J Mol Sci 2025; 26:766. [PMID: 39859478 PMCID: PMC11766068 DOI: 10.3390/ijms26020766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
Molar incisor hypomineralization (MIH) is a developmental defect that affects the enamel tissue of permanent teeth. Clinicians may observe a range of opacities in the affected teeth, varying from white to creamy, yellow, and brown. Of particular interest is an etiology of MIH that has not been rigorously elucidated. Researchers believe that there are many potential etiological factors with strong genetic and/or epigenetic influence. The primary factors contributing to the risk of MIH development include specific medical conditions and circumstances. These encompass prematurity, cesarean delivery, perinatal hypoxia, and various health issues such as measles, urinary tract infections, otitis media, gastrointestinal disorders, bronchitis, kidney diseases, pneumonia, and asthma. Although genetic research in this area has received substantial attention, the investigation of epigenetic factors remains comparatively underexplored. Special attention is given to genes and their protein products involved in amelogenesis. Examples of such genes are AMELX, AMBN, ENAM, TUFT1, FAM83H, and MMP20. The median relative FAM83H gene expression in the control group was 0.038 (0.031-0.061) and 0.045 (0.032-0.087) in the study group in buccal swabs. The median relative TUFT1 gene expression in the control group was 0.328 (0.247-0.456) and 0.704 (0.334-1.183) in the study group in buccal swabs. Furthermore, children with MIH had significantly higher TUFT1 expression levels compared to the control group (p-value = 0.0043). Alterations in the expression of the TUFT1 and FAM83H genes could be contributing factors to MIH pathogenesis. Nonetheless, further investigation is essential to comprehensively elucidate the roles of all analyzed genes in the pathogenesis of MIH.
Collapse
Affiliation(s)
- Wojciech Tynior
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Dorota Hudy
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Karolina Gołąbek
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Agnieszka Raczkowska-Siostrzonek
- Department of Dental Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 17 Plac Akademicki, 41-902 Bytom, Poland
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| |
Collapse
|
4
|
Jones RA, Cooper F, Kelly G, Barry D, Renshaw MJ, Sapkota G, Smith JC. Zebrafish reveal new roles for Fam83f in hatching and the DNA damage-mediated autophagic response. Open Biol 2024; 14:240194. [PMID: 39437839 PMCID: PMC11495952 DOI: 10.1098/rsob.240194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 10/25/2024] Open
Abstract
The FAM83 (Family with sequence similarity 83) family is highly conserved in vertebrates, but little is known of the functions of these proteins beyond their association with oncogenesis. Of the family, FAM83F is of particular interest because it is the only membrane-targeted FAM83 protein. When overexpressed, FAM83F activates the canonical Wnt signalling pathway and binds to and stabilizes p53; it therefore interacts with two pathways often dysregulated in disease. Insights into gene function can often be gained by studying the roles they play during development, and here we report the generation of fam83f knock-out (KO) zebrafish, which we have used to study the role of Fam83f in vivo. We show that endogenous fam83f is most strongly expressed in the hatching gland of developing zebrafish embryos, and that fam83f KO embryos hatch earlier than their wild-type (WT) counterparts, despite developing at a comparable rate. We also demonstrate that fam83f KO embryos are more sensitive to ionizing radiation than WT embryos-an unexpected finding, bearing in mind the previously reported ability of FAM83F to stabilize p53. Transcriptomic analysis shows that loss of fam83f leads to downregulation of phosphatidylinositol-3-phosphate (PI(3)P) binding proteins and impairment of cellular degradation pathways, particularly autophagy, a crucial component of the DNA damage response. Finally, we show that Fam83f protein is itself targeted to the lysosome when overexpressed in HEK293T cells, and that this localization is dependent upon a C' terminal signal sequence. The zebrafish lines we have generated suggest that Fam83f plays an important role in autophagic/lysosomal processes, resulting in dysregulated hatching and increased sensitivity to genotoxic stress in vivo.
Collapse
Affiliation(s)
- Rebecca A. Jones
- Department of Molecular Biology, Princeton University, Princeton, NJ08544, USA
| | - Fay Cooper
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, UK
- Neuroscience Institute, University of Sheffield, SheffieldS10 2TN, UK
| | - Gavin Kelly
- The Francis Crick Institute, 1 Midland Road, LondonNW1 1AT, UK
| | - David Barry
- The Francis Crick Institute, 1 Midland Road, LondonNW1 1AT, UK
| | | | - Gopal Sapkota
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, DundeeDD1 5EH, UK
| | - James C. Smith
- The Francis Crick Institute, 1 Midland Road, LondonNW1 1AT, UK
| |
Collapse
|
5
|
White MJ, Jacobs KA, Singh T, Mayo LN, Lin A, Chen CS, Jun YW, Kutys ML. Notch1 cortical signaling regulates epithelial architecture and cell-cell adhesion. J Cell Biol 2023; 222:e202303013. [PMID: 37796194 PMCID: PMC10555887 DOI: 10.1083/jcb.202303013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 10/06/2023] Open
Abstract
Notch receptors control tissue morphogenic processes that involve coordinated changes in cell architecture and gene expression, but how a single receptor can produce these diverse biological outputs is unclear. Here, we employ a 3D model of a human ductal epithelium to reveal tissue morphogenic defects result from loss of Notch1, but not Notch1 transcriptional signaling. Instead, defects in duct morphogenesis are driven by dysregulated epithelial cell architecture and mitogenic signaling which result from the loss of a transcription-independent, Notch1 cortical signaling mechanism that ultimately functions to stabilize adherens junctions and cortical actin. We identify that Notch1 localization and cortical signaling are tied to apical-basal cell restructuring and discover that a Notch1-FAM83H interaction underlies control of epithelial adherens junctions and cortical actin. Together, these results offer new insights into Notch1 signaling and regulation and advance a paradigm in which transcriptional and cell adhesive programs might be coordinated by a single receptor.
Collapse
Affiliation(s)
- Matthew J. White
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - Kyle A. Jacobs
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Tania Singh
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
| | - Lakyn N. Mayo
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
| | - Annie Lin
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
- Department of Otolaryngology, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Christopher S. Chen
- Department of Biomedical Engineering, The Biological Design Center, Boston University, Boston, MA, USA
| | - Young-wook Jun
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
- Department of Otolaryngology, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Matthew L. Kutys
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
6
|
Dong J, Ruan W, Duan X. Molecular-based phenotype variations in amelogenesis imperfecta. Oral Dis 2023; 29:2334-2365. [PMID: 37154292 DOI: 10.1111/odi.14599] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 04/03/2023] [Accepted: 04/15/2023] [Indexed: 05/10/2023]
Abstract
Amelogenesis imperfecta (AI) is one of the typical dental genetic diseases in human. It can occur isolatedly or as part of a syndrome. Previous reports have mainly clarified the types and mechanisms of nonsyndromic AI. This review aimed to compare the phenotypic differences among the hereditary enamel defects with or without syndromes and their underlying pathogenic genes. We searched the articles in PubMed with different strategies or keywords including but not limited to amelogenesis imperfecta, enamel defects, hypoplastic/hypomaturation/hypocalcified, syndrome, or specific syndrome name. The articles with detailed clinical information about the enamel and other phenotypes and clear genetic background were used for the analysis. We totally summarized and compared enamel phenotypes of 18 nonsyndromic AI with 17 causative genes and 19 syndromic AI with 26 causative genes. According to the clinical features, radiographic or ultrastructural changes in enamel, the enamel defects were basically divided into hypoplastic and hypomineralized (hypomaturated and hypocalcified) and presented a higher heterogeneity which were closely related to the involved pathogenic genes, types of mutation, hereditary pattern, X chromosome inactivation, incomplete penetrance, and other mechanisms.The gene-specific enamel phenotypes could be an important indicator for diagnosing nonsyndromic and syndromic AI.
Collapse
Affiliation(s)
- Jing Dong
- State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Diseases, School of Stomatology, National Clinical Research Center for Oral Disease, The Fourth Military Medical University, Xi'an, China
- College of Life Sciences, Northwest University, Xi'an, China
| | - Wenyan Ruan
- State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Diseases, School of Stomatology, National Clinical Research Center for Oral Disease, The Fourth Military Medical University, Xi'an, China
| | - Xiaohong Duan
- State Key Laboratory of Military Stomatology, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Diseases, School of Stomatology, National Clinical Research Center for Oral Disease, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
7
|
Bloch-Zupan A, Rey T, Jimenez-Armijo A, Kawczynski M, Kharouf N, O-Rare consortium, Dure-Molla MDL, Noirrit E, Hernandez M, Joseph-Beaudin C, Lopez S, Tardieu C, Thivichon-Prince B, ERN Cranio Consortium, Dostalova T, Macek M, International Consortium, Alloussi ME, Qebibo L, Morkmued S, Pungchanchaikul P, Orellana BU, Manière MC, Gérard B, Bugueno IM, Laugel-Haushalter V. Amelogenesis imperfecta: Next-generation sequencing sheds light on Witkop's classification. Front Physiol 2023; 14:1130175. [PMID: 37228816 PMCID: PMC10205041 DOI: 10.3389/fphys.2023.1130175] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/06/2023] [Indexed: 05/27/2023] Open
Abstract
Amelogenesis imperfecta (AI) is a heterogeneous group of genetic rare diseases disrupting enamel development (Smith et al., Front Physiol, 2017a, 8, 333). The clinical enamel phenotypes can be described as hypoplastic, hypomineralized or hypomature and serve as a basis, together with the mode of inheritance, to Witkop's classification (Witkop, J Oral Pathol, 1988, 17, 547-553). AI can be described in isolation or associated with others symptoms in syndromes. Its occurrence was estimated to range from 1/700 to 1/14,000. More than 70 genes have currently been identified as causative. Objectives: We analyzed using next-generation sequencing (NGS) a heterogeneous cohort of AI patients in order to determine the molecular etiology of AI and to improve diagnosis and disease management. Methods: Individuals presenting with so called "isolated" or syndromic AI were enrolled and examined at the Reference Centre for Rare Oral and Dental Diseases (O-Rares) using D4/phenodent protocol (www.phenodent.org). Families gave written informed consents for both phenotyping and molecular analysis and diagnosis using a dedicated NGS panel named GenoDENT. This panel explores currently simultaneously 567 genes. The study is registered under NCT01746121 and NCT02397824 (https://clinicaltrials.gov/). Results: GenoDENT obtained a 60% diagnostic rate. We reported genetics results for 221 persons divided between 115 AI index cases and their 106 associated relatives from a total of 111 families. From this index cohort, 73% were diagnosed with non-syndromic amelogenesis imperfecta and 27% with syndromic amelogenesis imperfecta. Each individual was classified according to the AI phenotype. Type I hypoplastic AI represented 61 individuals (53%), Type II hypomature AI affected 31 individuals (27%), Type III hypomineralized AI was diagnosed in 18 individuals (16%) and Type IV hypoplastic-hypomature AI with taurodontism concerned 5 individuals (4%). We validated the genetic diagnosis, with class 4 (likely pathogenic) or class 5 (pathogenic) variants, for 81% of the cohort, and identified candidate variants (variant of uncertain significance or VUS) for 19% of index cases. Among the 151 sequenced variants, 47 are newly reported and classified as class 4 or 5. The most frequently discovered genotypes were associated with MMP20 and FAM83H for isolated AI. FAM20A and LTBP3 genes were the most frequent genes identified for syndromic AI. Patients negative to the panel were resolved with exome sequencing elucidating for example the gene involved ie ACP4 or digenic inheritance. Conclusion: NGS GenoDENT panel is a validated and cost-efficient technique offering new perspectives to understand underlying molecular mechanisms of AI. Discovering variants in genes involved in syndromic AI (CNNM4, WDR72, FAM20A … ) transformed patient overall care. Unravelling the genetic basis of AI sheds light on Witkop's AI classification.
Collapse
Affiliation(s)
- Agnes Bloch-Zupan
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Université de Strasbourg, Institut d’études avancées (USIAS), Strasbourg, France
- Hôpitaux Universitaires de Strasbourg (HUS), Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpital Civil, Centre de référence des maladies rares orales et dentaires, O-Rares, Filiére Santé Maladies rares TETE COU, European Reference Network ERN CRANIO, Strasbourg, France
- Université de Strasbourg, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), IN-SERM U1258, CNRS- UMR7104, Illkirch, France
- Eastman Dental Institute, University College London, London, United Kingdom
| | - Tristan Rey
- Université de Strasbourg, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), IN-SERM U1258, CNRS- UMR7104, Illkirch, France
- Hôpitaux Universitaires de Strasbourg, Laboratoires de diagnostic génétique, Institut de Génétique Médicale d’Alsace, Strasbourg, France
| | - Alexandra Jimenez-Armijo
- Université de Strasbourg, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), IN-SERM U1258, CNRS- UMR7104, Illkirch, France
| | - Marzena Kawczynski
- Hôpitaux Universitaires de Strasbourg (HUS), Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpital Civil, Centre de référence des maladies rares orales et dentaires, O-Rares, Filiére Santé Maladies rares TETE COU, European Reference Network ERN CRANIO, Strasbourg, France
| | - Naji Kharouf
- Université de Strasbourg, Laboratoire de Biomatériaux et Bioingénierie, Inserm UMR_S 1121, Strasbourg, France
| | | | - Muriel de La Dure-Molla
- Rothschild Hospital, Public Assistance-Paris Hospitals (AP-HP), Reference Center for Rare Oral and Den-tal Diseases (O-Rares), Paris, France
| | - Emmanuelle Noirrit
- Centre Hospitalier Universitaire (CHU) Rangueil, Toulouse, Competence Center for Rare Oral and Den-tal Diseases, Toulouse, France
| | - Magali Hernandez
- Centre Hospitalier Régional Universitaire de Nancy, Université de Lorraine, Competence Center for Rare Oral and Dental Diseases, Nancy, France
| | - Clara Joseph-Beaudin
- Centre Hospitalier Universitaire de Nice, Competence Center for Rare Oral and Dental Diseases, Nice, France
| | - Serena Lopez
- Centre Hospitalier Universitaire de Nantes, Competence Center for Rare Oral and Dental Diseases, Nantes, France
| | - Corinne Tardieu
- APHM, Hôpitaux Universitaires de Marseille, Hôpital Timone, Competence Center for Rare Oral and Dental Diseases, Marseille, France
| | - Béatrice Thivichon-Prince
- Centre Hospitalier Universitaire de Lyon, Competence Center for Rare Oral and Dental Diseases, Lyon, France
| | | | - Tatjana Dostalova
- Department of Stomatology (TD) and Department of Biology and Medical Genetics (MM) Charles University 2nd Faculty of Medicine and Motol University Hospital, Prague, Czechia
| | - Milan Macek
- Department of Stomatology (TD) and Department of Biology and Medical Genetics (MM) Charles University 2nd Faculty of Medicine and Motol University Hospital, Prague, Czechia
| | | | - Mustapha El Alloussi
- Faculty of Dentistry, International University of Rabat, CReSS Centre de recherche en Sciences de la Santé, Rabat, Morocco
| | - Leila Qebibo
- Unité de génétique médicale et d’oncogénétique, CHU Hassan II, Fes, Morocco
| | | | | | - Blanca Urzúa Orellana
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Marie-Cécile Manière
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Hôpitaux Universitaires de Strasbourg (HUS), Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpital Civil, Centre de référence des maladies rares orales et dentaires, O-Rares, Filiére Santé Maladies rares TETE COU, European Reference Network ERN CRANIO, Strasbourg, France
| | - Bénédicte Gérard
- Hôpitaux Universitaires de Strasbourg, Laboratoires de diagnostic génétique, Institut de Génétique Médicale d’Alsace, Strasbourg, France
| | - Isaac Maximiliano Bugueno
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Hôpitaux Universitaires de Strasbourg (HUS), Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpital Civil, Centre de référence des maladies rares orales et dentaires, O-Rares, Filiére Santé Maladies rares TETE COU, European Reference Network ERN CRANIO, Strasbourg, France
- Université de Strasbourg, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), IN-SERM U1258, CNRS- UMR7104, Illkirch, France
| | - Virginie Laugel-Haushalter
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Université de Strasbourg, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), IN-SERM U1258, CNRS- UMR7104, Illkirch, France
- Hôpitaux Universitaires de Strasbourg, Laboratoires de diagnostic génétique, Institut de Génétique Médicale d’Alsace, Strasbourg, France
| |
Collapse
|
8
|
White MJ, Jacobs KA, Singh T, Kutys ML. Notch1 cortical signaling regulates epithelial architecture and cell-cell adhesion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.524428. [PMID: 36747830 PMCID: PMC9900753 DOI: 10.1101/2023.01.23.524428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Notch receptors control tissue morphogenic processes that involve coordinated changes in cell architecture and gene expression, but how a single receptor can produce these diverse biological outputs is unclear. Here we employ a 3D organotypic model of a ductal epithelium to reveal tissue morphogenic defects result from loss of Notch1, but not Notch1 transcriptional signaling. Instead, defects in duct morphogenesis are driven by dysregulated epithelial cell architecture and mitogenic signaling which result from loss of a transcription-independent Notch1 cortical signaling mechanism that ultimately functions to stabilize adherens junctions and cortical actin. We identify that Notch1 localization and cortical signaling are tied to apical-basal cell restructuring and discover a Notch1-FAM83H interaction underlies stabilization of adherens junctions and cortical actin. Together, these results offer new insights into Notch1 signaling and regulation, and advance a paradigm in which transcriptional and cell adhesive programs might be coordinated by a single receptor.
Collapse
Affiliation(s)
- Matthew J. White
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco CA, 94143, USA
| | - Kyle A. Jacobs
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco CA, 94143, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco CA, 94143, USA
| | - Tania Singh
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco CA, 94143, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco, University of California Berkeley, San Francisco CA, 94143, USA
| | - Matthew L. Kutys
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco CA, 94143, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco CA, 94143, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco, University of California Berkeley, San Francisco CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco CA, 94143, USA
| |
Collapse
|
9
|
Zheng X, Huang W, He Z, Li Y, Li S, Song Y. Effects of Fam83h truncation mutation on enamel developmental defects in male C57/BL6J mice. Bone 2023; 166:116595. [PMID: 36272714 DOI: 10.1016/j.bone.2022.116595] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/10/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022]
Abstract
Truncation mutations in family with sequence similarity, member H (FAM83H) gene are considered the main cause of autosomal dominant hypocalcified amelogenesis imperfecta (ADHCAI); however, its pathogenic mechanism in amelogenesis remains poorly characterized. This study aimed to investigate the effects of truncated FAM83H on developmental defects in enamel. CRISPR/Cas9 technology was used to develop a novel Fam83h c.1186C > T (p.Q396*) knock-in mouse strain, homologous to the human FAM83H c.1192C > T mutation in ADHCAI. The Fam83hQ396⁎/Q396⁎ mice showed poor growth, a sparse and scruffy coat, scaly skin and early mortality compared to control mice. Moreover, the forelimbs of homozygous mice were swollen, exhibiting a significant inflammatory response. Incisors of Fam83hQ396⁎/Q396⁎ mice appeared chalky white, shorter, and less sharp than those of control mice, and energy dispersive X-ray spectroscopy (EDS) analysis and Prussian blue staining helped identify decreased iron and increased calcium (Ca) and phosphorus (P) levels, with an unchanged Ca/P ratio. The expression of iron transportation proteins, transferrin receptor (TFRC) and solute carrier family 40 member 1 (SLC40A1), was decreased in Fam83h-mutated ameloblasts. Micro-computed tomography revealed enamel defects in Fam83hQ396⁎/Q396⁎ mice. Fam83hQ396⁎/Q396⁎ enamel showed decreased Vickers hardness and distorted enamel rod structure and ameloblast arrangement. mRNA sequencing showed that the cell adhesion pathway was most notably clustered in LS8-Fam83h-mutated cells. Immunofluorescence analysis further revealed decreased protein expression of desmoglein 3, a component of desmosomes, in Fam83h-mutated ameloblasts. The FAM83H-casein kinase 1α (CK1α)-keratin 14 (K14)-amelogenin (AMELX) interaction was detected in ameloblasts. And K14 and AMELX were disintegrated from the tetramer in Fam83h-mutated ameloblasts in vitro and in vivo. In secretory stage ameloblasts of Fam83hQ396⁎/Q396⁎ mice, AMELX secretion exhibited obvious retention in the cytoplasm. In conclusion, truncated FAM83H exerted dominant-negative effects on gross development, amelogenesis, and enamel biomineralization by disturbing iron transportation, influencing the transportation and secretion of AMELX, and interfering with cell-cell adhesion in ameloblasts.
Collapse
Affiliation(s)
- Xueqing Zheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wushuang Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhenru He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shiyu Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yaling Song
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei_MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
10
|
Rao MMV, Likith M, Kavya R, Hariprasad TPN. Plectin as a putative novel biomarker for breast cancer: an in silico study. NETWORK MODELING ANALYSIS IN HEALTH INFORMATICS AND BIOINFORMATICS 2022; 11:49. [DOI: 10.1007/s13721-022-00392-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 10/06/2022] [Accepted: 10/26/2022] [Indexed: 01/03/2025]
|
11
|
Zhang Y, Yang J, Yao H, Zhang Z, Song Y. CRISPR
/Cas9‐mediated deletion of
Fam83h
induces defective tooth mineralization and hair development in rabbits. J Cell Mol Med 2022; 26:5670-5679. [DOI: 10.1111/jcmm.17597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 08/16/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Yuxin Zhang
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University Changchun China
| | - Jie Yang
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University Changchun China
| | - Haobin Yao
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University Changchun China
| | - Zhongtian Zhang
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University Changchun China
| | - Yuning Song
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University Changchun China
| |
Collapse
|
12
|
The conserved C-terminal residues of FAM83H are required for the recruitment of casein kinase 1 to the keratin cytoskeleton. Sci Rep 2022; 12:11819. [PMID: 35821396 PMCID: PMC9276658 DOI: 10.1038/s41598-022-16153-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
The casein kinase 1 (CK1) family of serine/threonine protein kinases is involved in diverse cellular events at discrete subcellular compartments. FAM83H acts as a scaffold protein that recruits CK1 to the keratin cytoskeleton or to the nuclear speckles, which are storage sites for splicing factors. We determined the amino acid region of FAM83H required for recruiting CK1 to the keratin cytoskeleton. The subcellular localization of mutant FAM83H proteins with deletions of amino acid residues at different positions was evaluated via immunofluorescence. FAM83H mutants with deleted C-terminal residues 1134–1139, which are conserved among vertebrates, lost the ability to localize and recruit CK1 to the keratin cytoskeleton, suggesting that these residues are required for recruiting CK1 to the keratin cytoskeleton. The deletion of these residues (1134–1139) translocated FAM83H and CK1 to the nuclear speckles. Amino acid residues 1 to 603 of FAM83H were determined to contain the region responsible for the recruitment of CK1 to the nuclear speckles. Our results indicated that FAM83H recruits CK1 preferentially to the keratin cytoskeleton and alternatively to the nuclear speckles.
Collapse
|
13
|
Hunziker A, Glas I, Pohl MO, Stertz S. Phosphoproteomic profiling of influenza virus entry reveals infection-triggered filopodia induction counteracted by dynamic cortactin phosphorylation. Cell Rep 2022; 38:110306. [DOI: 10.1016/j.celrep.2022.110306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/19/2021] [Accepted: 01/06/2022] [Indexed: 11/03/2022] Open
|
14
|
Tokuchi K, Kitamura S, Maeda T, Watanabe M, Hatakeyama S, Kano S, Tanaka S, Ujiie H, Yanagi T. Loss of FAM83H promotes cell migration and invasion in cutaneous squamous cell carcinoma via impaired keratin distribution. J Dermatol Sci 2021; 104:112-121. [PMID: 34657752 DOI: 10.1016/j.jdermsci.2021.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUNDS FAM83H is essential for amelogenesis, but recent reports implicate that FAM83H is involved in the tumorigenesis. We previously clarified that TRIM29 binds to FAM83H to regulate keratin distribution and squamous cell migration. However, little is known about FAM83H in normal/malignant skin keratinocytes. OBJECTIVE To investigate the expression of FAM83H in cutaneous squamous cell carcinoma (SCC) and its physiological function. METHODS Immunohistochemical analysis and RT-PCR of human SCC tissues were performed. Next, we examined the effect of FAM83H knockdown/overexpression in SCC cell lines using cell proliferation, migration, and invasion assay. To investigate the molecular mechanism, immunoprecipitation of FAM83H was examined. Further, Immunofluorescence staining was performed. Finally, we examined the correlation between the expressions of FAM83H and the keratin distribution. RESULTS FAM83H expression was lower in SCC lesions than in normal epidermis and correlated with differentiation grade. The mRNA expression levels of FAM83H in SCC tumors were also lower than in normal epidermis. The knockdown of FAM83H enhanced SCC cell migration and invasion, whereas the overexpression of FAM83H led to decreases in both. Furthermore, the knockdown of FAM83H enhanced the cancer cell metastasis in vivo. FAM83H formed a complex with TRIM29 and keratins. The knockdown of FAM83H altered keratin distribution and solubility. Clinically, the loss of FAM83H correlates with an altered keratin distribution. CONCLUSION Our findings reveal a critical function for FAM83H in regulating keratin distribution, as well as in the migration/invasion of cutaneous SCC, suggesting that FAM83H could be a crucial molecule in the tumorigenesis of cutaneous SCC.
Collapse
Affiliation(s)
- Keiko Tokuchi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| | - Shinya Kitamura
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| | - Takuya Maeda
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| | - Masashi Watanabe
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| | - Shigetsugu Hatakeyama
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| | - Satoshi Kano
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine and WPI-ICReDD, Hokkaido University, Japan
| | - Hideyuki Ujiie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| | - Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| |
Collapse
|
15
|
Fulcher LJ, Sapkota GP. Functions and regulation of the serine/threonine protein kinase CK1 family: moving beyond promiscuity. Biochem J 2020; 477:4603-4621. [PMID: 33306089 PMCID: PMC7733671 DOI: 10.1042/bcj20200506] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
Regarded as constitutively active enzymes, known to participate in many, diverse biological processes, the intracellular regulation bestowed on the CK1 family of serine/threonine protein kinases is critically important, yet poorly understood. Here, we provide an overview of the known CK1-dependent cellular functions and review the emerging roles of CK1-regulating proteins in these processes. We go on to discuss the advances, limitations and pitfalls that CK1 researchers encounter when attempting to define relationships between CK1 isoforms and their substrates, and the challenges associated with ascertaining the correct physiological CK1 isoform for the substrate of interest. With increasing interest in CK1 isoforms as therapeutic targets, methods of selectively inhibiting CK1 isoform-specific processes is warranted, yet challenging to achieve given their participation in such a vast plethora of signalling pathways. Here, we discuss how one might shut down CK1-specific processes, without impacting other aspects of CK1 biology.
Collapse
Affiliation(s)
- Luke J. Fulcher
- Department of Biochemistry, University of Oxford, Oxford, U.K
| | - Gopal P. Sapkota
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, U.K
| |
Collapse
|
16
|
Leal-Gutiérrez JD, Elzo MA, Carr C, Mateescu RG. RNA-seq analysis identifies cytoskeletal structural genes and pathways for meat quality in beef. PLoS One 2020; 15:e0240895. [PMID: 33175867 PMCID: PMC7657496 DOI: 10.1371/journal.pone.0240895] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 10/05/2020] [Indexed: 01/03/2023] Open
Abstract
RNA sequencing (RNA-seq) has allowed for transcriptional profiling of biological systems through the identification of differentially expressed (DE) genes and pathways. A total of 80 steers with extreme phenotypes were selected from the University of Florida multibreed Angus-Brahman herd. The average slaughter age was 12.91±8.69 months. Tenderness, juiciness and connective tissue assessed by sensory panel, along with marbling, Warner-Bratzler Shear Force (WBSF) and cooking loss, were measured in longissimus dorsi muscle. Total RNA was extracted from muscle and one RNA-seq library per sample was constructed, multiplexed, and sequenced based on protocols by Illumina HiSeq-3000 platform to generate 2×101 bp paired-end reads. The overall read mapping rate using the Btau_4.6.1 reference genome was 63%. A total of 8,799 genes were analyzed using two different methodologies, an expression association and a DE analysis. A gene and exon expression association analysis was carried out using a meat quality index on all 80 samples as a continuous response variable. The expression of 208 genes and 3,280 exons from 1,565 genes was associated with the meat quality index (p-value ≤ 0.05). A gene and isoform DE evaluation was performed analyzing two groups with extreme WBSF, tenderness and marbling. A total of 676 (adjusted p-value≤0.05), 70 (adjusted p-value≤0.1) and 198 (adjusted p-value≤0.1) genes were DE for WBSF, tenderness and marbling, respectively. A total of 106 isoforms from 98 genes for WBSF, 13 isoforms from 13 genes for tenderness and 43 isoforms from 42 genes for marbling (FDR≤0.1) were DE. Cytoskeletal and transmembrane anchoring genes and pathways were identified in the expression association, DE and the gene enrichment analyses; these proteins can have a direct effect on meat quality. Cytoskeletal proteins and transmembrane anchoring molecules can influence meat quality by allowing cytoskeletal interaction with myocyte and organelle membranes, contributing to cytoskeletal structure and architecture maintenance postmortem.
Collapse
Affiliation(s)
- Joel D. Leal-Gutiérrez
- Department of Animal Sciences, University of Florida, Gainesville, Florida, United States of America
| | - Mauricio A. Elzo
- Department of Animal Sciences, University of Florida, Gainesville, Florida, United States of America
| | - Chad Carr
- Department of Animal Sciences, University of Florida, Gainesville, Florida, United States of America
| | - Raluca G. Mateescu
- Department of Animal Sciences, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
17
|
Wang SK, Zhang H, Hu CY, Liu JF, Chadha S, Kim JW, Simmer JP, Hu JCC. FAM83H and Autosomal Dominant Hypocalcified Amelogenesis Imperfecta. J Dent Res 2020; 100:293-301. [PMID: 33034243 DOI: 10.1177/0022034520962731] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Autosomal dominant hypocalcified amelogenesis imperfecta (ADHCAI; OMIM #130900) is a genetic disorder exhibiting severe hardness defects and reduced fracture toughness of dental enamel. While the condition is nonsyndromic, it can be associated with other craniofacial anomalies, such as malocclusions and delayed or failed tooth eruption. Truncation mutations in FAM83H (OMIM *611927) are hitherto the sole cause of ADHCAI. With human genetic studies, Fam83h knockout and mutation-knock-in mouse models indicated that FAM83H does not serve a critical physiologic function during enamel formation and suggested a neomorphic mutation mechanism causing ADHCAI. The function of FAM83H remains obscure. FAM83H has been shown to interact with various isoforms of casein kinase 1 (CK1) and keratins and to mediate organization of keratin cytoskeletons and desmosomes. By considering FAM83H a scaffold protein to anchor CK1s, further molecular characterization of the protein could gain insight into its functions. In this study, we characterized 9 kindreds with ADHCAI and identified 3 novel FAM83H truncation mutations: p.His437*, p.Gln459*, and p.Glu610*. Some affected individuals exhibited hypoplastic phenotypes, in addition to the characteristic hypocalcification enamel defects, which have never been well documented. Failed eruption of canines or second molars in affected persons was observed in 4 of the families. The p.Glu610* mutation was located in a gap area (amino acids 470 to 625) within the zone of previously reported pathogenic variants (amino acids 287 to 694). In vitro pull-down studies with overexpressed FAM83H proteins in HEK293 cells demonstrated an interaction between FAM83H and SEC16A, a protein component of the COP II complex at endoplasmic reticulum exit sites. The interaction was mediated by the middle part (amino acids 287 to 657) of mouse FAM83H protein. Results of this study significantly extended the phenotypic and genotypic spectrums of FAM83H-associated ADHCAI and suggested a role for FAM83H in endoplasmic reticulum-to-Golgi vesicle trafficking and protein secretion (dbGaP phs001491.v1.p1).
Collapse
Affiliation(s)
- S K Wang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.,Department of Dentistry, School of Dentistry, National Taiwan University, Jhongjheng District, Taipei City, Taiwan
| | - H Zhang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - C Y Hu
- Department of Prosthodontics, National Taiwan University Hospital, Jhongjheng District, Taipei City, Taiwan
| | - J F Liu
- Division of Pediatric Dentistry, Department of Stomatology, Taichung Veterans General Hospital, Xitun District, Taichung City, Taiwan
| | - S Chadha
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - J W Kim
- Department of Pediatric Dentistry and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea.,Department of Molecular Genetics and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - J P Simmer
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - J C C Hu
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
18
|
Ishikawa T, Terashima J, Sasaki D, Shimoyama Y, Yaegashi T, Sasaki M. Establishment and use of a three-dimensional ameloblastoma culture model to study the effects of butyric acid on the transcription of growth factors and laminin β3. Arch Oral Biol 2020; 118:104845. [PMID: 32712305 DOI: 10.1016/j.archoralbio.2020.104845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE This study aimed to establish a three-dimensional (3D) culture method for ameloblastoma cell lines and to use the model to investigate the effect of butyric acid (BA), a periodontopathic bacterial metabolite, on the malignant transformation of ameloblastoma. DESIGN Three ameloblastoma cell lines (HAM1, HAM2, and HAM3) established from the same tumor were used in this study. A 3D culture model was established in low absorption dishes and was incubated for 48 h. The effects of BA on the transcription of growth factors and LMβ3 were examined by real-time reverse transcription PCR. Various BA concentrations (0.02, 0.2, 2, and 20 mM) were used to stimulate the cell cultures for 6 and 12 h. RESULTS A 3D culture model was established. Gene expression levels of epithelial growth factor (EGF), transforming growth factor beta 1 (TGFβ1), and laminin β3 (LMβ3) were higher in 3D than in 2D cultures. Cell morphology in 3D cultures did not change, while the transcription levels of EGF, TGFβ1, and LMβ3 were upregulated by BA in all cell lines. CONCLUSION The 3D culture model is more responsive to BA than the 2D culture model, and there is a possibility that the malignancy and progression of ameloblastoma via laminin 332 (LM332) is mediated by BA.
Collapse
Affiliation(s)
- Taichi Ishikawa
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University, 1-1-1 Idai-dori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3694, Japan.
| | - Jun Terashima
- Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University, 1-1-1 Idai-dori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3694, Japan
| | - Daisuke Sasaki
- Division of Periodontology, Department of Conservative Dentistry, School of Dentistry, Iwate Medical University, 1-3-27 Chuo-dori, Morioka, Iwate, 020-8505, Japan
| | - Yu Shimoyama
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University, 1-1-1 Idai-dori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3694, Japan
| | - Takashi Yaegashi
- Division of Periodontology, Department of Conservative Dentistry, School of Dentistry, Iwate Medical University, 1-3-27 Chuo-dori, Morioka, Iwate, 020-8505, Japan
| | - Minoru Sasaki
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University, 1-1-1 Idai-dori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3694, Japan
| |
Collapse
|
19
|
Ishikawa T, Terashima J, Shimoyama Y, Ohashi Y, Mikami T, Takeda Y, Sasaki M. Effects of butyric acid, a bacterial metabolite, on the migration of ameloblastoma mediated by laminin 332. J Oral Sci 2020; 62:435-438. [PMID: 32879156 DOI: 10.2334/josnusd.19-0380] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Ameloblastoma is a benign tumor that develops in the jawbone. Occasionally, however, it may become malignant and metastasize to other tissues. Although it has been suggested that various cytokines and several adhesion factors may play a role in its malignant transformation, the details have not been elucidated. In this context, it has been reported that butyric acid produced by periodontopathic bacteria causes progression of malignant tumors occurring in the mouth via podoplanin. However, the influence of butyric acid on ameloblastoma has not been clarified. In the present study, therefore, the expression of various cytokines and adhesion factors in ameloblastoma upon stimulation with butyric acid or cytokines was investigated using real-time reverse-transcription polymerase chain reaction. Three cell lines (HAM1, HAM2 and HAM3) established from the same ameloblastoma were used in the experiments. It was found that the expression of mRNAs for epidermal growth factor (EGF) and transforming growth factor beta 1 (TGFβ1) was increased in HAM2 and HAM3, respectively, upon stimulation with butyric acid. In addition, stimulation with EGF and TGFβ1 led to an increase in the expression of laminin β-3 mRNA in the respective cell lines. These results suggest that butyric acid may be involved in ameloblastoma exacerbation through the expression of laminin 332 (LM332) via EGF and TGFβ1 produced by ameloblastoma itself.
Collapse
Affiliation(s)
- Taichi Ishikawa
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University
| | - Jun Terashima
- Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University
| | - Yu Shimoyama
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University
| | - Yu Ohashi
- Division of Oral and Maxillofacial Surgery, Department of Reconstructive Oral and Maxillofacial Surgery, School of Dentistry, Iwate Medical University
| | | | - Yasunori Takeda
- Division of Clinical Pathology, Department of Oral and Maxillofacial Reconstructive Surgery, School of Dentistry, Iwate Medical University
| | - Minoru Sasaki
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University
| |
Collapse
|
20
|
Expression of FAM83H and ZNF16 are associated with shorter survival of patients with gallbladder carcinoma. Diagn Pathol 2020; 15:63. [PMID: 32460791 PMCID: PMC7254718 DOI: 10.1186/s13000-020-00985-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/20/2020] [Indexed: 02/03/2023] Open
Abstract
Background Recently, FAM83H was reported to have roles in cancer progression in conjunction with oncogenic molecules such as MYC and b-catenin. Moreover, the data from the public database indicates a molecular relationship between FAM83H and zinc finger proteins, especially between FAM83H and ZNF16. However, studies on FAM83H and ZNF16 in gallbladder cancer have been limited. Methods This study investigated the expression of FAM83H and ZNF16 in 105 gallbladder carcinomas. Results In human gallbladder carcinomas, immunohistochemical expression of FAM83H was significantly associated with ZNF16 expression. In univariate analysis, nuclear and cytoplasmic expression of FAM83H or ZNF16 were significantly associated with shorter survival of gallbladder carcinoma patients. Multivariate analysis revealed the nuclear expression of FAM83H as an independent indicator of poor prognosis of overall survival (p = 0.005) and relapse-free survival (p = 0.005) of gallbladder carcinoma patients. Moreover, co-expression patterns of nuclear FAM83H and ZNF16 were also independent indicators of shorter survival of gallbladder carcinoma patients (overall survival; p < 0.001, relapse-free survival; p < 0.001). Conclusions This study suggests FAM83H and ZNF16 are associated with the progression of gallbladder carcinoma, and the expressions of FAM83H and ZNF16 might be novel prognostic indicators of gallbladder carcinoma patients.
Collapse
|
21
|
Zhuang H, Zhang C, Hou B. FAM83H overexpression predicts worse prognosis and correlates with less CD8 + T cells infiltration and Ras-PI3K-Akt-mTOR signaling pathway in pancreatic cancer. Clin Transl Oncol 2020; 22:2244-2252. [PMID: 32424701 DOI: 10.1007/s12094-020-02365-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 04/28/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND Family with sequence similarity 83 members H (FAM83H) is one member of Family with sequence similarity 83 (FAM83) family, which possess oncogenic properties in several types of cancer. However, the potential function of FAM83H in pancreatic cancer (PC) still remain unknown. AIM This study aims to explore the role of FAM83H during pancreatic carcinogenesis and the regulation of immune infiltration in PC. METHODS In the current study, the clinical significance and potential biological of FAM83H were evaluated by bioinformatics analysis. Possible associations between FAM83H expression and tumor immunity were analyzed using ESTIMATE algorithm and single-sample gene set enrichment analysis (ssGSEA). RESULTS FAM83H expression was significantly upregulated in tumor tissues, and positively associated with higher histologic grade, tumor recurrence, and worse prognosis. FAM83H overexpression is notably associated with KRAS activation. And functional enrichment analysis demonstrated that FAM83H may be involved in positive regulation of cell proliferation and migration, Ras protein signal transduction, regulation of cell-matrix adhesion, epithelial to mesenchymal transition (EMT), TGF-β receptor signaling in EMT, and activated NOTCH transmits signal to the nucleus. ESTIMATE algorithm and ssGSEA demonstrated that FAM83H overexpression suppressed the infiltration and antitumor activity of tumor-infiltrating lymphocytes (TILs), especially for CD8+ T cells. Besides, FAM83H overexpression significantly correlated with low expression of TIL-related gene markers (e.g. CD8A, CD8B, CD2, CD3D, and CD3E). CONCLUSION The study suggests that FAM83H overexpression predicts poor prognosis and correlates with less CD8+ T cells infiltration and Ras-PI3K-Akt-mTOR signaling pathway in PC.
Collapse
Affiliation(s)
- H Zhuang
- Shantou University of Medical College, Shantou, 515000, China
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - C Zhang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106 Zhongshan Er Road, Guangzhou, 510080, China.
| | - B Hou
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106 Zhongshan Er Road, Guangzhou, 510080, China.
| |
Collapse
|
22
|
Nowwarote N, Osathanon T, Kanjana K, Theerapanon T, Porntaveetus T, Shotelersuk V. Decreased osteogenic activity and mineralization of alveolar bone cells from a patient with amelogenesis imperfecta and FAM83H 1261G>T mutation. Genes Dis 2019; 6:391-397. [PMID: 31832519 PMCID: PMC6889029 DOI: 10.1016/j.gendis.2019.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/10/2019] [Accepted: 07/18/2019] [Indexed: 12/27/2022] Open
Abstract
FAM83H mutations lead to autosomal dominant hypocalcified amelogenesis imperfecta (ADHCAI). However, the biological role of FAM83H remains unclear. The present study aimed to characterize the alveolar bone cells isolated from a patient with ADHCAI having the mutation, c.1261G > T, p.E421*, in FAM83H. We showed that FAM83H mutant cells had proliferation ability and morphology similar to the controls. The F-actin staining revealed that FAM83H mutant cells were remained in the earlier stages of cell spreading compared to the controls at 30 min, but their spreading was advanced comparable to the controls at later stages. After osteogenic induction, a significant decrease in mRNA levels of RUNX2 and ALP was observed in FAM83H mutant cells at day 7 compared with day 3 while their expressions were increased in the controls. The OPN levels in FAM83H mutant cells were not significantly changed at day 7 compared to day 3 while the controls showed a significant increase. After 14 days, the mineral deposition of FAM83H mutant cells was slightly lower than that of the controls. In conclusion, we identify that FAM83H bone cells have lower expression of osteogenic marker genes and mineralization while they maintain their morphology, proliferation, and spreading. Consistent with previous studies in the ameloblasts and periodontal ligamental cells, these evidences propose that FAM83H influences osteogenic differentiation across different cell types in oral cavity.
Collapse
Affiliation(s)
- Nunthawan Nowwarote
- Genomics and Precision Dentistry Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanaphum Osathanon
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kiattipan Kanjana
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanakorn Theerapanon
- Genomics and Precision Dentistry Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thantrira Porntaveetus
- Genomics and Precision Dentistry Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Corresponding author. Genomics and Precision Dentistry Research Unit, Faculty of Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand. Fax: +662 218 8691.
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Excellence Center for Medical Genetics, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, 10330, Thailand
| |
Collapse
|
23
|
Nasseri S, Nikkho B, Parsa S, Ebadifar A, Soleimani F, Rahimi K, Vahabzadeh Z, Khadem-Erfan MB, Rostamzadeh J, Baban B, Banafshi O, Assadollahi V, Mirzaie S, Fathi F. Generation of Fam83h knockout mice by CRISPR/Cas9-mediated gene engineering. J Cell Biochem 2019; 120:11033-11043. [PMID: 30714208 DOI: 10.1002/jcb.28381] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 12/19/2018] [Accepted: 01/07/2019] [Indexed: 01/24/2023]
Abstract
Family with sequence similarity 83 member H (FAM83H) protein-coding geneplay an essential role in the structural organization, calcification of developing enamel, and keratin cytoskeleton disassembly by recruiting Casein kinase 1 alpha (CSNK1A1) to keratin filaments. In this study, we have applied CRISPR Cas9 nickase (D10A) to knockout (KO) the Fam83h gene in NMRI outbred mice. We generated homozygous Fam83h KO mice ( Fam83h Ko/Ko ) through a premature termination codon, which was validated by Sanger sequencing in F0 generation. Next, we also bred the FAM83H KO for two generations. Reverse-transcription polymerase chain reaction and Western blot analysis approved the Fam83h KO mice. The Fam83h KO mice had evidence of normal morphology at the cervical loops, secretory and maturation stages, and mandibular molars. In comparison with the normal wild-type mice ( Fam83h W/W ), the F2 homozygous KO ( Fam83h Ko/Ko ) had sparse, scruffy coats with small body size and decreased general activity. Also, they had the natural reproductive ability and natural lifespan. In addition, delay in opening the eyes and dry eyes among infant mice were seen. The F1 heterozygous mice looked comparable to the normal wild-type mice ( Fam83h W/W ), which showed autosomal recessive inheritance of these phenotypes. The KO of FAM83H had controversial effects on the development of teeth and the formation of enamel. The phenotype defect in dental development and the enamel formation were seen in three mice among four generations. It can be concluded that null FAM83H in outbred mice not only showed the reported phenotypes in null inbred mouse but also showed normal lifespan and reproductive ability; dental deficiency in three homozygous mice; and the symptoms that were similar to the symptoms of dry eye syndrome and curly coat dog syndrome in all four evaluated KO generations.
Collapse
Affiliation(s)
- Sherko Nasseri
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Bahram Nikkho
- Department of Pathology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sara Parsa
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Asghar Ebadifar
- Dentofacial Deformities Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Soleimani
- Department of Biology, School of Natural Science, University of Tabriz, Tabriz, Iran
| | - Karim Rahimi
- Department of Molecular Biology and Genetics-Gene Expression and Gene Medicine, Aarhus University, Aarhus, Denmark
| | - Zakaria Vahabzadeh
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Bagher Khadem-Erfan
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jalal Rostamzadeh
- Department of Animal Science, Faculty of Agriculture, University of Kurdistan, Sanandaj, Iran
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia
| | - Omid Banafshi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Vahideh Assadollahi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sako Mirzaie
- Department of Biochemistry, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Fardin Fathi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
24
|
Chen C, Li HF, Hu YJ, Jiang MJ, Liu QS, Zhou J. Family with Sequence Similarity 83 Member H Promotes the Viability and Metastasis of Cervical Cancer Cells and Indicates a Poor Prognosis. Yonsei Med J 2019; 60:611-618. [PMID: 31250574 PMCID: PMC6597464 DOI: 10.3349/ymj.2019.60.7.611] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 04/22/2019] [Accepted: 05/09/2019] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Family with sequence similarity 83 member H (FAM83H) plays key roles in tumorigenesis. However, the specific roles of FAM83H in cervical cancer (CC) have not been well studied. MATERIALS AND METHODS The RNA-seq data of 306 CC tissues and three normal samples downloaded from The Cancer Genome Atlas were used to analyze the expression of FAM83H. The Kaplan-Meier method was used to draw survival curves. Associations between FAM83H expression and clinicopathological factors were analyzed by chi-square test. Cox proportional hazards model was used to analyze prognostic factors. Loss-of-function assays were conducted to discover the biological functions of FAM83H in cell proliferation, colony formation, invasion, and migration. Real-time Quantitative Reverse Transcription PCR (qRT-PCR) and Western blotting were used to measure the expression levels of FAM83H in CC cell lines. RESULTS Our results demonstrated that FAM83H is overexpressed in CC tissues and that high FAM83H expression is associated with worse overall survival (OS). High FAM83H expression in CC was associated with clinical stage, pathologic tumor, and pathologic node. Univariate analysis suggested that FAM83H expression was significantly related to the OS of CC patients. Although multivariate analysis showed that FAM83H expression was not an independent prognostic factor for the OS of CC patients, the effects of FAM83H on CC cell growth and motility was significant. Loss-of-function experiments demonstrated that knockdown of FAM83H inhibited proliferation, colony formation, migration, and invasion of CC cells by inactivating PI3K/AKT pathway. CONCLUSION FAM83H might play a crucial role in CC progression and could act as a novel therapeutic target in CC.
Collapse
Affiliation(s)
- Chao Chen
- Department of Radiotherapy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Hua Feng Li
- Department of Radiotherapy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Yu Jie Hu
- Department of Radiotherapy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Meng Jie Jiang
- Department of Radiotherapy, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Qing Sheng Liu
- Department of Geratology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Jia Zhou
- Department of Geratology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
25
|
Long non-coding RNA FAM83H-AS1 is regulated by human papillomavirus 16 E6 independently of p53 in cervical cancer cells. Sci Rep 2019; 9:3662. [PMID: 30842470 PMCID: PMC6403315 DOI: 10.1038/s41598-019-40094-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 12/20/2018] [Indexed: 12/30/2022] Open
Abstract
High-risk human papillomavirus (HPV) infection is one of the first events in the process of carcinogenesis in cervical and head and neck cancers. The expression of the viral oncoproteins E6 and E7 are essential in this process by inactivating the tumor suppressor proteins p53 and Rb, respectively, in addition to their interactions with other host proteins. Non-coding RNAs, such as long non-coding RNAs (lncRNAs) have been found to be dysregulated in several cancers, suggesting an important role in tumorigenesis. In order to identify host lncRNAs affected by HPV infection, we expressed the high-risk HPV-16 E6 oncoprotein in primary human keratinocytes and measured the global lncRNA expression profile by high-throughput sequencing (RNA-seq). We found several host lncRNAs differentially expressed by E6 including GAS5, H19, and FAM83H-AS1. Interestingly, FAM83H-AS1 was found overexpressed in HPV-16 positive cervical cancer cell lines in an HPV-16 E6-dependent manner but independently of p53 regulation. Furthermore, FAM83H-AS1 was found to be regulated through the E6-p300 pathway. Knockdown of FAM83H-AS1 by siRNAs decreased cellular proliferation, migration and increased apoptosis. FAM83H-AS1 was also found to be altered in human cervical cancer tissues and high expression of this lncRNA was associated with worse overall survival, suggesting an important role in cervical carcinogenesis.
Collapse
|
26
|
Arnes L, Liu Z, Wang J, Maurer HC, Sagalovskiy I, Sanchez-Martin M, Bommakanti N, Garofalo DC, Balderes DA, Sussel L, Olive KP, Rabadan R. Comprehensive characterisation of compartment-specific long non-coding RNAs associated with pancreatic ductal adenocarcinoma. Gut 2019; 68:499-511. [PMID: 29440233 PMCID: PMC6086768 DOI: 10.1136/gutjnl-2017-314353] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 12/22/2017] [Accepted: 01/01/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDA) is a highly metastatic disease with limited therapeutic options. Genome and transcriptome analyses have identified signalling pathways and cancer driver genes with implications in patient stratification and targeted therapy. However, these analyses were performed in bulk samples and focused on coding genes, which represent a small fraction of the genome. DESIGN We developed a computational framework to reconstruct the non-coding transcriptome from cross-sectional RNA-Seq, integrating somatic copy number alterations (SCNA), common germline variants associated to PDA risk and clinical outcome. We validated the results in an independent cohort of paired epithelial and stromal RNA-Seq derived from laser capture microdissected human pancreatic tumours, allowing us to annotate the compartment specificity of their expression. We employed systems and experimental biology approaches to interrogate the function of epithelial long non-coding RNAs (lncRNAs) associated with genetic traits and clinical outcome in PDA. RESULTS We generated a catalogue of PDA-associated lncRNAs. We showed that lncRNAs define molecular subtypes with biological and clinical significance. We identified lncRNAs in genomic regions with SCNA and single nucleotide polymorphisms associated with lifetime risk of PDA and associated with clinical outcome using genomic and clinical data in PDA. Systems biology and experimental functional analysis of two epithelial lncRNAs (LINC00673 and FAM83H-AS1) suggest they regulate the transcriptional profile of pancreatic tumour samples and PDA cell lines. CONCLUSIONS Our findings indicate that lncRNAs are associated with genetic marks of pancreatic cancer risk, contribute to the transcriptional regulation of neoplastic cells and provide an important resource to design functional studies of lncRNAs in PDA.
Collapse
Affiliation(s)
- Luis Arnes
- Columbia University Medical Center, New York, United States
- Department of Biomedical Informatics, Hong Kong University of Science and Technology, Hong Kong
- Department of Systems Biology, Hong Kong University of Science and Technology, Hong Kong
| | - Zhaoqi Liu
- Columbia University Medical Center, New York, United States
- Department of Biomedical Informatics, Hong Kong University of Science and Technology, Hong Kong
- Department of Systems Biology, Hong Kong University of Science and Technology, Hong Kong
| | - Jiguang Wang
- Columbia University Medical Center, New York, United States
- Department of Biomedical Informatics, Hong Kong University of Science and Technology, Hong Kong
- Division of Life Science and Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong
| | - H. Carlo Maurer
- Columbia University Medical Center, New York, United States
- Department of Medicine, Division of Digestive and Liver Diseases, University of Colorado, Denver, United States
| | - Irina Sagalovskiy
- Columbia University Medical Center, New York, United States
- Department of Biomedical Informatics, Hong Kong University of Science and Technology, Hong Kong
- Department of Systems Biology, Hong Kong University of Science and Technology, Hong Kong
| | - Marta Sanchez-Martin
- Columbia University Medical Center, New York, United States
- Institute for Cancer Genetics, University of Colorado, Denver, United States
| | - Nikhil Bommakanti
- Columbia University Medical Center, New York, United States
- Department of Biomedical Informatics, Hong Kong University of Science and Technology, Hong Kong
- Department of Systems Biology, Hong Kong University of Science and Technology, Hong Kong
| | - Diana C. Garofalo
- Columbia University Medical Center, New York, United States
- Department of Genetics & Development, University of Colorado, Denver, United States
| | - Dina A. Balderes
- Columbia University Medical Center, New York, United States
- Department of Genetics & Development, University of Colorado, Denver, United States
| | - Lori Sussel
- Columbia University Medical Center, New York, United States
- Department of Genetics & Development, University of Colorado, Denver, United States
- Barbara Davis Center, University of Colorado, Denver, United States
| | - Kenneth P. Olive
- Columbia University Medical Center, New York, United States
- Department of Medicine, Division of Digestive and Liver Diseases, University of Colorado, Denver, United States
- Department of Pathology and Cell Biology
- Herbert Irving Comprehensive Cancer Center
| | - Raul Rabadan
- Columbia University Medical Center, New York, United States
- Department of Biomedical Informatics, Hong Kong University of Science and Technology, Hong Kong
- Department of Systems Biology, Hong Kong University of Science and Technology, Hong Kong
| |
Collapse
|
27
|
Kim KM, Hussein UK, Bae JS, Park SH, Kwon KS, Ha SH, Park HS, Lee H, Chung MJ, Moon WS, Kang MJ, Jang KY. The Expression Patterns of FAM83H and PANX2 Are Associated With Shorter Survival of Clear Cell Renal Cell Carcinoma Patients. Front Oncol 2019; 9:14. [PMID: 30723706 PMCID: PMC6349742 DOI: 10.3389/fonc.2019.00014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 01/04/2019] [Indexed: 12/21/2022] Open
Abstract
FAM83H is primarily known for its role in amelogenesis; however, recent reports suggest FAM83H might be involved in tumorigenesis. Although the studies of FAM83H in kidney cancer are limited, a search of the public database shows a significant association between FAM83H and pannexin-2 (PANX2) in clear cell renal cell carcinomas (CCRCCs). Therefore, we evaluated the clinicopathological significance of the immunohistochemical expression of FAM83H and PANX2 in 199 CCRCC patients. The expression of FAM83H and PANX2 were significantly associated with each other. In univariate analysis, individual, and co-expression pattern of FAM83H and PANX2 was significantly associated with shorter overall survival (OS) and relapse-free survival (RFS) of CCRCC patients: nuclear expression of FAM83H (OS; P < 0.001, RFS; P < 0.001), cytoplasmic expression of FAM83H (OS; P < 0.001, RFS; P < 0.001), nuclear expression of PANX2 (OS; P < 0.001, RFS; P < 0.001), cytoplasmic expression of PANX2 (OS; P < 0.001, RFS; P < 0.001), co-expression pattern of nuclear FAM83H and nuclear PANX2 (OS; P < 0.001, RFS; P < 0.001). In multivariate analysis, nuclear expression of FAM83H (OS; P < 0.001, RFS; P = 0.003) and the co-expression pattern of nuclear FAM83H and PANX2 (OS; P < 0.001, RFS; P < 0.001) were independent indicators of shorter survival of CCRCC patients. Cytoplasmic expression of FAM83H was associated with shorter RFS (P = 0.030) in multivariate analysis. In Caki-1 and Caki-2 CCRCC cells, knock-down of FAM83H decreased PANX2 expression and cell proliferation, and overexpression of FAM83H increased PANX2 expression and cell proliferation. These results suggest that FAM83H and PANX2 might be involved in the progression of CCRCC in a co-operative manner, and their expression might be used as novel prognostic indicators for CCRCC patients.
Collapse
Affiliation(s)
- Kyoung Min Kim
- Department of Pathology, Chonbuk National University Medical School, Jeonju, South Korea.,Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, South Korea.,Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju, South Korea
| | - Usama Khamis Hussein
- Department of Pathology, Chonbuk National University Medical School, Jeonju, South Korea.,Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Jun Sang Bae
- Department of Pathology, Chonbuk National University Medical School, Jeonju, South Korea.,Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, South Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Keun Sang Kwon
- Department of Preventive Medicine, Chonbuk National University Medical School, Jeonju, South Korea
| | - Sang Hoon Ha
- Division of Biotechnology, Chonbuk National University, Iksan, South Korea
| | - Ho Sung Park
- Department of Pathology, Chonbuk National University Medical School, Jeonju, South Korea.,Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, South Korea
| | - Ho Lee
- Department of Forensic Medicine, Chonbuk National University Medical School, Jeonju, South Korea
| | - Myoung Ja Chung
- Department of Pathology, Chonbuk National University Medical School, Jeonju, South Korea.,Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, South Korea
| | - Woo Sung Moon
- Department of Pathology, Chonbuk National University Medical School, Jeonju, South Korea.,Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, South Korea
| | - Myoung Jae Kang
- Department of Pathology, Chonbuk National University Medical School, Jeonju, South Korea.,Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, South Korea
| | - Kyu Yun Jang
- Department of Pathology, Chonbuk National University Medical School, Jeonju, South Korea.,Research Institute of Clinical Medicine of Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, South Korea.,Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju, South Korea
| |
Collapse
|
28
|
Yanagi T, Watanabe M, Hata H, Kitamura S, Imafuku K, Yanagi H, Homma A, Wang L, Takahashi H, Shimizu H, Hatakeyama S. Loss of TRIM29 Alters Keratin Distribution to Promote Cell Invasion in Squamous Cell Carcinoma. Cancer Res 2018; 78:6795-6806. [PMID: 30389700 DOI: 10.1158/0008-5472.can-18-1495] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/28/2018] [Accepted: 10/10/2018] [Indexed: 11/16/2022]
Abstract
: TRIM29 (tripartite motif-containing protein 29) is a TRIM family protein that has been implicated in breast, colorectal, and pancreatic cancers. However, its role in stratified squamous epithelial cells and tumors has not been elucidated. Here, we investigate the expression of TRIM29 in cutaneous head and neck squamous cell carcinomas (SCC) and its functions in the tumorigenesis of such cancers. TRIM29 expression was lower in malignant SCC lesions than in adjacent normal epithelial tissue or benign tumors. Lower expression of TRIM29 was associated with higher SCC invasiveness. Primary tumors of cutaneous SCC showed aberrant hypermethylation of TRIM29. Depletion of TRIM29 increased cancer cell migration and invasion; conversely, overexpression of TRIM29 suppressed these. Comprehensive proteomics and immunoprecipitation analyses identified keratins and keratin-interacting protein FAM83H as TRIM29 interactors. Knockdown of TRIM29 led to ectopic keratin localization of keratinocytes. In primary tumors, lower TRIM29 expression correlated with the altered expression of keratins. Our findings reveal an unexpected role for TRIM29 in regulating the distribution of keratins, as well as in the migration and invasion of SCC. They also suggest that the TRIM29-keratin axis could serve as a diagnostic and prognostic marker in stratified epithelial tumors and may provide a target for SCC therapeutics. SIGNIFICANCE: These findings identify TRIM29 as a novel diagnostic and prognostic marker in stratified epithelial tissues.
Collapse
Affiliation(s)
- Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Masashi Watanabe
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Hiroo Hata
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shinya Kitamura
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Keisuke Imafuku
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroko Yanagi
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Akihiro Homma
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Hokkaido, Japan
| | - Hidehisa Takahashi
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, Yokohama, Japan
| | - Hiroshi Shimizu
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shigetsugu Hatakeyama
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan.
| |
Collapse
|
29
|
Nowwarote N, Theerapanon T, Osathanon T, Pavasant P, Porntaveetus T, Shotelersuk V. Amelogenesis imperfecta: A novel FAM83H mutation and characteristics of periodontal ligament cells. Oral Dis 2018; 24:1522-1531. [PMID: 29949226 DOI: 10.1111/odi.12926] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/14/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To delineate orodental features, dental mineral density, genetic aetiology and cellular characteristics associated with amelogenesis imperfecta (AI). MATERIALS AND METHODS Three affected patients in a family were recruited. Whole-exome sequencing was used to identify mutations confirmed by Sanger sequencing. The proband's teeth were subjected for mineral density analysis by microcomputerised tomography and characterisation of periodontal ligament cells (PDLCs). RESULTS The patients presented yellow-brown, pitted and irregular enamel. A novel nonsense mutation, c.1261G>T, p.E421*, in exon 5 of the FAM83H was identified. The mineral density of the enamel was significantly decreased in the proband. The patient's PDLCs (FAM83H cells) exhibited reduced ability of cell proliferation and colony-forming unit compared with controls. The formation of stress fibres was remarkably present. Upon cultured in osteogenic induction medium, FAM83H cells, at day 7 compared to day 3, had a significant reduction of BSP, COL1 and OCN mRNA expression and no significant change in RUNX2. The upregulation of ALP mRNA levels and mineral deposition were comparable between FAM83H and control cells. CONCLUSIONS We identified the novel mutation in FAM83H associated with autosomal dominant hypocalcified AI. The FAM83H cells showed reduced cell proliferation and expression of osteogenic markers, suggesting altered PDLCs in FAM83H-associated AI.
Collapse
Affiliation(s)
- Nunthawan Nowwarote
- Genomics and Precision Dentistry Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanakorn Theerapanon
- Genomics and Precision Dentistry Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Excellence Center in Regenerative Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Excellence Center in Regenerative Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thantrira Porntaveetus
- Genomics and Precision Dentistry Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Excellence Center for Medical Genetics, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
30
|
The FAM83 family of proteins: from pseudo-PLDs to anchors for CK1 isoforms. Biochem Soc Trans 2018; 46:761-771. [PMID: 29871876 PMCID: PMC6008594 DOI: 10.1042/bst20160277] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
Abstract
The eight members of the FAM83 (FAMily with sequence similarity 83) family of poorly characterised proteins are only present in vertebrates and are defined by the presence of the conserved DUF1669 domain of unknown function at their N-termini. The DUF1669 domain consists of a conserved phospholipase D (PLD)-like catalytic motif. However, the FAM83 proteins display no PLD catalytic (PLDc) activity, and the pseudo-PLDc motif present in each FAM83 member lacks the crucial elements of the native PLDc motif. In the absence of catalytic activity, it is likely that the DUF1669 domain has evolved to espouse novel function(s) in biology. Recent studies have indicated that the DUF1669 domain mediates the interaction with different isoforms of the CK1 (casein kinase 1) family of Ser/Thr protein kinases. In turn, different FAM83 proteins, which exhibit unique amino acid sequences outside the DUF1669 domain, deliver CK1 isoforms to unique subcellular compartments. One of the first protein kinases to be discovered, the CK1 isoforms are thought to be constitutively active and are known to control a plethora of biological processes. Yet, their regulation of kinase activity, substrate selectivity and subcellular localisation has remained a mystery. The emerging evidence now supports a central role for the DUF1669 domain, and the FAM83 proteins, in the regulation of CK1 biology.
Collapse
|
31
|
Yang M, Huang W, Yang F, Zhang T, Wang C, Song Y. Fam83h mutation inhibits the mineralization in ameloblasts by activating Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2018; 501:206-211. [DOI: 10.1016/j.bbrc.2018.04.216] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 01/27/2023]
|
32
|
Fulcher LJ, Bozatzi P, Tachie-Menson T, Wu KZL, Cummins TD, Bufton JC, Pinkas DM, Dunbar K, Shrestha S, Wood NT, Weidlich S, Macartney TJ, Varghese J, Gourlay R, Campbell DG, Dingwell KS, Smith JC, Bullock AN, Sapkota GP. The DUF1669 domain of FAM83 family proteins anchor casein kinase 1 isoforms. Sci Signal 2018; 11:eaao2341. [PMID: 29789297 PMCID: PMC6025793 DOI: 10.1126/scisignal.aao2341] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Members of the casein kinase 1 (CK1) family of serine-threonine protein kinases are implicated in the regulation of many cellular processes, including the cell cycle, circadian rhythms, and Wnt and Hedgehog signaling. Because these kinases exhibit constitutive activity in biochemical assays, it is likely that their activity in cells is controlled by subcellular localization, interactions with inhibitory proteins, targeted degradation, or combinations of these mechanisms. We identified members of the FAM83 family of proteins as partners of CK1 in cells. All eight members of the FAM83 family (FAM83A to FAM83H) interacted with the α and α-like isoforms of CK1; FAM83A, FAM83B, FAM83E, and FAM83H also interacted with the δ and ε isoforms of CK1. We detected no interaction between any FAM83 member and the related CK1γ1, CK1γ2, and CK1γ3 isoforms. Each FAM83 protein exhibited a distinct pattern of subcellular distribution and colocalized with the CK1 isoform(s) to which it bound. The interaction of FAM83 proteins with CK1 isoforms was mediated by the conserved domain of unknown function 1669 (DUF1669) that characterizes the FAM83 family. Mutations in FAM83 proteins that prevented them from binding to CK1 interfered with the proper subcellular localization and cellular functions of both the FAM83 proteins and their CK1 binding partners. On the basis of its function, we propose that DUF1669 be renamed the polypeptide anchor of CK1 domain.
Collapse
Affiliation(s)
- Luke J Fulcher
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Polyxeni Bozatzi
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Theresa Tachie-Menson
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Kevin Z L Wu
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Timothy D Cummins
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Joshua C Bufton
- Structural Genomics Consortium, University of Oxford, Oxford, UK
| | - Daniel M Pinkas
- Structural Genomics Consortium, University of Oxford, Oxford, UK
| | - Karen Dunbar
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Sabin Shrestha
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Nicola T Wood
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Simone Weidlich
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Thomas J Macartney
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Joby Varghese
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - Robert Gourlay
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | - David G Campbell
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK
| | | | | | - Alex N Bullock
- Structural Genomics Consortium, University of Oxford, Oxford, UK
| | - Gopal P Sapkota
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, Dundee, Scotland, UK.
| |
Collapse
|
33
|
Schlögl E, Radeva MY, Vielmuth F, Schinner C, Waschke J, Spindler V. Keratin Retraction and Desmoglein3 Internalization Independently Contribute to Autoantibody-Induced Cell Dissociation in Pemphigus Vulgaris. Front Immunol 2018; 9:858. [PMID: 29922278 PMCID: PMC5996934 DOI: 10.3389/fimmu.2018.00858] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/06/2018] [Indexed: 01/12/2023] Open
Abstract
Pemphigus vulgaris (PV) is a potentially lethal autoimmune disease characterized by blister formation of the skin and mucous membranes and is caused by autoantibodies against desmoglein (Dsg) 1 and Dsg3. Dsg1 and Dsg3 are linked to keratin filaments in desmosomes, adhering junctions abundant in tissues exposed to high levels of mechanical stress. The binding of the autoantibodies leads to internalization of Dsg3 and a collapse of the keratin cytoskeleton-yet, the relevance and interdependence of these changes for loss of cell-cell adhesion and blistering is poorly understood. In live-cell imaging studies, loss of the keratin network at the cell periphery was detectable starting after 60 min of incubation with immunoglobulin G fractions of PV patients (PV-IgG). These rapid changes correlated with loss of cell-cell adhesion detected by dispase-based dissociation assays and were followed by a condensation of keratin filaments into thick bundles after several hours. Dsg3 internalization started at 90 min of PV-IgG treatment, thus following the early keratin changes. By inhibiting casein kinase 1 (CK-1), we provoked keratin alterations resembling the effects of PV-IgG. Although CK-1-induced loss of peripheral keratin network correlated with loss of cell cohesion and Dsg3 clustering in the membrane, it was not sufficient to trigger the internalization of Dsg3. However, additional incubation with PV-IgG was effective to promote Dsg3 loss at the membrane, indicating that Dsg3 internalization is independent from keratin alterations. Vice versa, inhibiting Dsg3 internalization did not prevent PV-IgG-induced keratin retraction and only partially rescued cell cohesion. Together, keratin changes appear very early after autoantibody binding and temporally overlap with loss of cell cohesion. These early alterations appear to be distinct from Dsg3 internalization, suggesting a crucial role for initial loss of cell cohesion in PV.
Collapse
Affiliation(s)
- Elisabeth Schlögl
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Franziska Vielmuth
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Camilla Schinner
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Volker Spindler
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany.,Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
34
|
Nishikawa S. Cytoskeleton, intercellular junctions, planar cell polarity, and cell movement in amelogenesis. J Oral Biosci 2017. [DOI: 10.1016/j.job.2017.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
35
|
Lacruz RS, Habelitz S, Wright JT, Paine ML. DENTAL ENAMEL FORMATION AND IMPLICATIONS FOR ORAL HEALTH AND DISEASE. Physiol Rev 2017; 97:939-993. [PMID: 28468833 DOI: 10.1152/physrev.00030.2016] [Citation(s) in RCA: 287] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/10/2017] [Accepted: 01/10/2017] [Indexed: 12/16/2022] Open
Abstract
Dental enamel is the hardest and most mineralized tissue in extinct and extant vertebrate species and provides maximum durability that allows teeth to function as weapons and/or tools as well as for food processing. Enamel development and mineralization is an intricate process tightly regulated by cells of the enamel organ called ameloblasts. These heavily polarized cells form a monolayer around the developing enamel tissue and move as a single forming front in specified directions as they lay down a proteinaceous matrix that serves as a template for crystal growth. Ameloblasts maintain intercellular connections creating a semi-permeable barrier that at one end (basal/proximal) receives nutrients and ions from blood vessels, and at the opposite end (secretory/apical/distal) forms extracellular crystals within specified pH conditions. In this unique environment, ameloblasts orchestrate crystal growth via multiple cellular activities including modulating the transport of minerals and ions, pH regulation, proteolysis, and endocytosis. In many vertebrates, the bulk of the enamel tissue volume is first formed and subsequently mineralized by these same cells as they retransform their morphology and function. Cell death by apoptosis and regression are the fates of many ameloblasts following enamel maturation, and what cells remain of the enamel organ are shed during tooth eruption, or are incorporated into the tooth's epithelial attachment to the oral gingiva. In this review, we examine key aspects of dental enamel formation, from its developmental genesis to the ever-increasing wealth of data on the mechanisms mediating ionic transport, as well as the clinical outcomes resulting from abnormal ameloblast function.
Collapse
Affiliation(s)
- Rodrigo S Lacruz
- Department of Basic Science and Craniofacial Biology, College of Dentistry, New York University, New York, New York; Department of Preventive and Restorative Dental Sciences, University of California, San Francisco, San Francisco, California; Department of Pediatric Dentistry, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina; Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California
| | - Stefan Habelitz
- Department of Basic Science and Craniofacial Biology, College of Dentistry, New York University, New York, New York; Department of Preventive and Restorative Dental Sciences, University of California, San Francisco, San Francisco, California; Department of Pediatric Dentistry, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina; Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California
| | - J Timothy Wright
- Department of Basic Science and Craniofacial Biology, College of Dentistry, New York University, New York, New York; Department of Preventive and Restorative Dental Sciences, University of California, San Francisco, San Francisco, California; Department of Pediatric Dentistry, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina; Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California
| | - Michael L Paine
- Department of Basic Science and Craniofacial Biology, College of Dentistry, New York University, New York, New York; Department of Preventive and Restorative Dental Sciences, University of California, San Francisco, San Francisco, California; Department of Pediatric Dentistry, School of Dentistry, University of North Carolina, Chapel Hill, North Carolina; Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California
| |
Collapse
|
36
|
Balmer P, Bauer A, Pujar S, McGarvey KM, Welle M, Galichet A, Müller EJ, Pruitt KD, Leeb T, Jagannathan V. A curated catalog of canine and equine keratin genes. PLoS One 2017; 12:e0180359. [PMID: 28846680 PMCID: PMC5573215 DOI: 10.1371/journal.pone.0180359] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/14/2017] [Indexed: 01/03/2023] Open
Abstract
Keratins represent a large protein family with essential structural and functional roles in epithelial cells of skin, hair follicles, and other organs. During evolution the genes encoding keratins have undergone multiple rounds of duplication and humans have two clusters with a total of 55 functional keratin genes in their genomes. Due to the high similarity between different keratin paralogs and species-specific differences in gene content, the currently available keratin gene annotation in species with draft genome assemblies such as dog and horse is still imperfect. We compared the National Center for Biotechnology Information (NCBI) (dog annotation release 103, horse annotation release 101) and Ensembl (release 87) gene predictions for the canine and equine keratin gene clusters to RNA-seq data that were generated from adult skin of five dogs and two horses and from adult hair follicle tissue of one dog. Taking into consideration the knowledge on the conserved exon/intron structure of keratin genes, we annotated 61 putatively functional keratin genes in both the dog and horse, respectively. Subsequently, curators in the RefSeq group at NCBI reviewed their annotation of keratin genes in the dog and horse genomes (Annotation Release 104 and Annotation Release 102, respectively) and updated annotation and gene nomenclature of several keratin genes. The updates are now available in the NCBI Gene database (https://www.ncbi.nlm.nih.gov/gene).
Collapse
Affiliation(s)
- Pierre Balmer
- Division of Clinical Dermatology, Department of Clinical Veterinary Medicine, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Anina Bauer
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Genetics, Vetsuisse Faculty, University of Bern,Bern, Switzerland
| | - Shashikant Pujar
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States of America
| | - Kelly M. McGarvey
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States of America
| | - Monika Welle
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Arnaud Galichet
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Department of Clinical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland
| | - Eliane J. Müller
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Department of Clinical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland
- Clinic for Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Kim D. Pruitt
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States of America
| | - Tosso Leeb
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Genetics, Vetsuisse Faculty, University of Bern,Bern, Switzerland
| | - Vidhya Jagannathan
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Genetics, Vetsuisse Faculty, University of Bern,Bern, Switzerland
- * E-mail:
| |
Collapse
|
37
|
Xin W, Wenjun W, Man Q, Yuming Z. Novel FAM83H mutations in patients with amelogenesis imperfecta. Sci Rep 2017; 7:6075. [PMID: 28729668 PMCID: PMC5519741 DOI: 10.1038/s41598-017-05208-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/25/2017] [Indexed: 11/09/2022] Open
Abstract
Amelogenesis imperfecta (AI), characterized by a deficiency in the quantity and/or quality of dental enamel, is genetically heterogeneous and phenotypically variable. The most severe type, hypocalcified AI, is mostly caused by truncating mutations in the FAM83H gene. This study aimed to identify genetic mutations in four Chinese families with hypocalcified AI. We performed mutation analysis by sequencing the candidate FAM83H gene. Three novel mutations (c.931dupC, p.V311Rfs*13; c.1130_1131delinsAA, p.S377X; and c.1147 G > T, p.E383X) and one previously reported mutation (c.973 C > T, p.R325X) in the last exon of FAM83H gene were identified. Furthermore, constructs expressing Green fluorescent protein (GFP)-tagged wild-type and three novel mutant FAM83Hs were transfected into rat dental epithelial cells (SF2 cells). Wild-type FAM83H-GFP was localized exclusively in the cytoplasm, especially in the area surrounding the nucleus, while the mutant FAM83H-GFPs (p.V311Rfs*13, p.S377X, and p.E383X) were localized predominantly in the nucleus, with lower levels in the cytoplasm.
Collapse
Affiliation(s)
- Wang Xin
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Wang Wenjun
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, PR China.,Beijing Key Laboratory of Digital Stomatology, Beijing, PR China
| | - Qin Man
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Zhao Yuming
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China.
| |
Collapse
|
38
|
Huang W, Yang M, Wang C, Song Y. Evolutionary analysis of FAM83H in vertebrates. PLoS One 2017; 12:e0180360. [PMID: 28683132 PMCID: PMC5500323 DOI: 10.1371/journal.pone.0180360] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/14/2017] [Indexed: 01/08/2023] Open
Abstract
Amelogenesis imperfecta is a group of disorders causing abnormalities in enamel formation in various phenotypes. Many mutations in the FAM83H gene have been identified to result in autosomal dominant hypocalcified amelogenesis imperfecta in different populations. However, the structure and function of FAM83H and its pathological mechanism have yet to be further explored. Evolutionary analysis is an alternative for revealing residues or motifs that are important for protein function. In the present study, we chose 50 vertebrate species in public databases representative of approximately 230 million years of evolution, including 1 amphibian, 2 fishes, 7 sauropsidas and 40 mammals, and we performed evolutionary analysis on the FAM83H protein. By sequence alignment, conserved residues and motifs were indicated, and the loss of important residues and motifs of five special species (Malayan pangolin, platypus, minke whale, nine-banded armadillo and aardvark) was discovered. A phylogenetic time tree showed the FAM83H divergent process. Positive selection sites in the C-terminus suggested that the C-terminus of FAM83H played certain adaptive roles during evolution. The results confirmed some important motifs reported in previous findings and identified some new highly conserved residues and motifs that need further investigation. The results suggest that the C-terminus of FAM83H contain key conserved regions critical to enamel formation and calcification.
Collapse
Affiliation(s)
- Wushuang Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mei Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Changning Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yaling Song
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- * E-mail:
| |
Collapse
|
39
|
Smith CEL, Poulter JA, Antanaviciute A, Kirkham J, Brookes SJ, Inglehearn CF, Mighell AJ. Amelogenesis Imperfecta; Genes, Proteins, and Pathways. Front Physiol 2017; 8:435. [PMID: 28694781 PMCID: PMC5483479 DOI: 10.3389/fphys.2017.00435] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/08/2017] [Indexed: 01/11/2023] Open
Abstract
Amelogenesis imperfecta (AI) is the name given to a heterogeneous group of conditions characterized by inherited developmental enamel defects. AI enamel is abnormally thin, soft, fragile, pitted and/or badly discolored, with poor function and aesthetics, causing patients problems such as early tooth loss, severe embarrassment, eating difficulties, and pain. It was first described separately from diseases of dentine nearly 80 years ago, but the underlying genetic and mechanistic basis of the condition is only now coming to light. Mutations in the gene AMELX, encoding an extracellular matrix protein secreted by ameloblasts during enamel formation, were first identified as a cause of AI in 1991. Since then, mutations in at least eighteen genes have been shown to cause AI presenting in isolation of other health problems, with many more implicated in syndromic AI. Some of the encoded proteins have well documented roles in amelogenesis, acting as enamel matrix proteins or the proteases that degrade them, cell adhesion molecules or regulators of calcium homeostasis. However, for others, function is less clear and further research is needed to understand the pathways and processes essential for the development of healthy enamel. Here, we review the genes and mutations underlying AI presenting in isolation of other health problems, the proteins they encode and knowledge of their roles in amelogenesis, combining evidence from human phenotypes, inheritance patterns, mouse models, and in vitro studies. An LOVD resource (http://dna2.leeds.ac.uk/LOVD/) containing all published gene mutations for AI presenting in isolation of other health problems is described. We use this resource to identify trends in the genes and mutations reported to cause AI in the 270 families for which molecular diagnoses have been reported by 23rd May 2017. Finally we discuss the potential value of the translation of AI genetics to clinical care with improved patient pathways and speculate on the possibility of novel treatments and prevention strategies for AI.
Collapse
Affiliation(s)
- Claire E L Smith
- Division of Oral Biology, School of Dentistry, St. James's University Hospital, University of LeedsLeeds, United Kingdom.,Section of Ophthalmology and Neuroscience, St. James's University Hospital, University of LeedsLeeds, United Kingdom
| | - James A Poulter
- Section of Ophthalmology and Neuroscience, St. James's University Hospital, University of LeedsLeeds, United Kingdom
| | - Agne Antanaviciute
- Section of Genetics, School of Medicine, St. James's University Hospital, University of LeedsLeeds, United Kingdom
| | - Jennifer Kirkham
- Division of Oral Biology, School of Dentistry, St. James's University Hospital, University of LeedsLeeds, United Kingdom
| | - Steven J Brookes
- Division of Oral Biology, School of Dentistry, St. James's University Hospital, University of LeedsLeeds, United Kingdom
| | - Chris F Inglehearn
- Section of Ophthalmology and Neuroscience, St. James's University Hospital, University of LeedsLeeds, United Kingdom
| | - Alan J Mighell
- Section of Ophthalmology and Neuroscience, St. James's University Hospital, University of LeedsLeeds, United Kingdom.,Oral Medicine, School of Dentistry, University of LeedsLeeds, United Kingdom
| |
Collapse
|
40
|
Kim KM, Park SH, Bae JS, Noh SJ, Tao GZ, Kim JR, Kwon KS, Park HS, Park BH, Lee H, Chung MJ, Moon WS, Sylvester KG, Jang KY. FAM83H is involved in the progression of hepatocellular carcinoma and is regulated by MYC. Sci Rep 2017; 7:3274. [PMID: 28607447 PMCID: PMC5468291 DOI: 10.1038/s41598-017-03639-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/02/2017] [Indexed: 01/25/2023] Open
Abstract
Recently, the roles of FAM83H in tumorigenesis have been interested and increased expression of FAM83H and MYC in hepatocellular carcinoma (HCC) have been reported. Therefore, we investigated the expression and role of FAM83H in 163 human HCCs and further investigated the relationship between FAM83H and oncogene MYC. The expression of FAM83H is elevated in liver cancer cells, and nuclear expression of FAM83H predicted shorter survival of HCC patients. In HLE and HepG2 HCC cells, knock-down of FAM83H inhibited proliferation and invasive activity of HCC cells. FAM83H induced expression of cyclin-D1, cyclin-E1, snail and MMP2 and inhibited the expression of P53 and P27. In hepatic tumor cells derived from Tet-O-MYC mice, the expression of mRNA and protein of FAM83H were dependent on MYC expression. Moreover, a chromatin immunoprecipitation assay demonstrated that MYC binds to the promotor of FAM83H and that MYC promotes the transcription of FAM83H, which was supported by the results of a dual-luciferase reporter assay. In conclusion, we present an oncogenic role of FAM83H in liver cancer, which is closely associated with the oncogene MYC. In addition, our results suggest FAM83H expression as a poor prognostic indicator of HCC patients.
Collapse
Affiliation(s)
- Kyoung Min Kim
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| | - Jun Sang Bae
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Sang Jae Noh
- Forensic Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Guo-Zhong Tao
- Department of Surgery, Division of Pediatric Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jung Ryul Kim
- Orthopedic Surgery, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Keun Sang Kwon
- Preventive Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Ho Sung Park
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Byung-Hyun Park
- Biochemistry, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Ho Lee
- Forensic Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Myoung Ja Chung
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Woo Sung Moon
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Karl G Sylvester
- Department of Surgery, Division of Pediatric Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Kyu Yun Jang
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea.
| |
Collapse
|
41
|
Hatzfeld M, Keil R, Magin TM. Desmosomes and Intermediate Filaments: Their Consequences for Tissue Mechanics. Cold Spring Harb Perspect Biol 2017; 9:a029157. [PMID: 28096266 PMCID: PMC5453391 DOI: 10.1101/cshperspect.a029157] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Adherens junctions (AJs) and desmosomes connect the actin and keratin filament networks of adjacent cells into a mechanical unit. Whereas AJs function in mechanosensing and in transducing mechanical forces between the plasma membrane and the actomyosin cytoskeleton, desmosomes and intermediate filaments (IFs) provide mechanical stability required to maintain tissue architecture and integrity when the tissues are exposed to mechanical stress. Desmosomes are essential for stable intercellular cohesion, whereas keratins determine cell mechanics but are not involved in generating tension. Here, we summarize the current knowledge of the role of IFs and desmosomes in tissue mechanics and discuss whether the desmosome-keratin scaffold might be actively involved in mechanosensing and in the conversion of chemical signals into mechanical strength.
Collapse
Affiliation(s)
- Mechthild Hatzfeld
- Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg, 06114 Halle, Germany
| | - René Keil
- Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg, 06114 Halle, Germany
| | - Thomas M Magin
- Institute of Biology, Division of Cell and Developmental Biology and Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
42
|
Snijders AM, Lee S, Hang B, Hao W, Bissell MJ, Mao J. FAM83 family oncogenes are broadly involved in human cancers: an integrative multi-omics approach. Mol Oncol 2017; 11:167-179. [PMID: 28078827 PMCID: PMC5527452 DOI: 10.1002/1878-0261.12016] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/16/2016] [Accepted: 10/21/2016] [Indexed: 01/15/2023] Open
Abstract
The development of novel targeted therapies for cancer treatment requires identification of reliable targets. FAM83 ('family with sequence similarity 83') family members A, B, and D were shown recently to have oncogenic potential. However, the overall oncogenic abilities of FAM83 family genes remain largely unknown. Here, we used a systematic and integrative genomics approach to investigate oncogenic properties of the entire FAM83 family members. We assessed transcriptional expression patterns of eight FAM83 family genes (FAM83A-H) across tumor types, the relationship between their expression and changes in DNA copy number, and the association with patient survival. By comparing the gene expression levels of FAM83 family members in cancers from 17 different tumor types with those in their corresponding normal tissues, we identified consistent upregulation of FAM83D and FAM83H across the majority of tumor types, which is largely driven by increased DNA copy number. Importantly, we found also that a higher expression level of a signature of FAM83 family members was associated with poor prognosis in a number of human cancers. In breast cancer, we found that alterations in FAM83 family genes correlated significantly with TP53 mutation, whereas significant, but inverse correlation was observed with PIK3CA and CDH1 (E-cadherin) mutations. We also identified that expression levels of 55 proteins were significantly associated with alterations in FAM83 family genes including a decrease in GATA3, ESR1, and PGR proteins in tumors with alterations in FAM83. Our results provide strong evidence for a critical role of FAM83 family genes in tumor development, with possible relevance for therapeutic target development.
Collapse
Affiliation(s)
- Antoine M. Snijders
- Biological Systems and Engineering DivisionLawrence Berkeley National LaboratoryCAUSA
| | - Sun‐Young Lee
- Biological Systems and Engineering DivisionLawrence Berkeley National LaboratoryCAUSA
| | - Bo Hang
- Biological Systems and Engineering DivisionLawrence Berkeley National LaboratoryCAUSA
| | - Wenshan Hao
- Nanjing Biotech and Pharmaceutical Valley Development CenterChina
| | - Mina J. Bissell
- Biological Systems and Engineering DivisionLawrence Berkeley National LaboratoryCAUSA
| | - Jian‐Hua Mao
- Biological Systems and Engineering DivisionLawrence Berkeley National LaboratoryCAUSA
| |
Collapse
|
43
|
Casein kinase 1 is recruited to nuclear speckles by FAM83H and SON. Sci Rep 2016; 6:34472. [PMID: 27681590 PMCID: PMC5041083 DOI: 10.1038/srep34472] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/14/2016] [Indexed: 11/29/2022] Open
Abstract
In some fibroblasts, casein kinase 1α (CK1α) is localized to nuclear speckles, which are sub-nuclear compartments supplying splicing factors, whereas it is recruited on keratin filaments in colorectal cancer cells such as DLD1 cells. In order to obtain a deeper understanding of why CK1α is localized to these different subcellular sites, we herein elucidated the mechanisms underlying its localization to nuclear speckles. CK1α and FAM83H were localized to nuclear speckles in RKO and WiDr colorectal cancer cells, which do not express simple epithelial keratins, and in DLD1 cells transfected with siRNAs for type I keratins. The localization of FAM83H to nuclear speckles was also detected in colorectal cancer cells with a poorly organized keratin cytoskeleton in colorectal cancer tissues. Using an interactome analysis of FAM83H, we identified SON, a protein present in nuclear speckles, as a scaffold protein to which FAM83H recruits CK1α. This result was supported by the knockdown of FAM83H or SON delocalizing CK1α from nuclear speckles. We also found that CK1δ and ε are localized to nuclear speckles in a FAM83H-dependent manner. These results suggest that CK1 is recruited to nuclear speckles by FAM83H and SON in the absence of an intact keratin cytoskeleton.
Collapse
|