1
|
Altunoglu U, Turgut GT, Özturan EK, Kalaycı T, Kaya M, Toksoy G, Baş F, Kayserili H, Darendeliler F. Expanding the Clinical and Mutational Spectrum of Biallelic POC1A Variants: Characterization of Four Patients and a Comprehensive Review of POC1A-Related Phenotypes. Clin Genet 2025; 107:390-401. [PMID: 39662966 DOI: 10.1111/cge.14672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
SOFT syndrome (SOFTS) is an autosomal recessive disorder caused by biallelic POC1A variants, characterized by short stature, distinctive facial features, onychodysplasia, and hypotrichosis. To date, 21 pathogenic POC1A variants have been reported in 26 families. This study aims to broaden the phenotypic and genotypic spectrum of SOFTS with emphasis on the long-term effects of growth hormone (GH) therapy. We report four unrelated patients with three homozygous POC1A variants and demonstrate the transcriptional effects of two canonical splicing variants. All four patients had severe growth retardation, sparse hair/eyebrows, high/prominent forehead, long/triangular face, prominent nose, short middle/distal phalanges, puffy/tapering fingers, and prominent heels. Endocrine abnormalities included insulin resistance and impaired glucose tolerance, dyslipidemia, GH deficiency, central hypothyroidism, and precocious puberty. Two patients treated long-term with recombinant human GH showed insufficient responses. We also provide an extensive review of 43 cases including those we report, contributing to a better understanding of the full clinical and endocrinological spectrum of SOFTS.
Collapse
Affiliation(s)
- Umut Altunoglu
- Medical Genetics Department, Koc University School of Medicine (KUSOM), Istanbul, Turkey
| | - Gozde Tutku Turgut
- Medical Genetics Department, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Esin Karakılıç Özturan
- Department of Pediatric Endocrinology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Tuğba Kalaycı
- Medical Genetics Department, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mert Kaya
- Graduate School of Health Sciences, Koc University, Istanbul, Turkey
| | - Güven Toksoy
- Medical Genetics Department, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Firdevs Baş
- Department of Pediatric Endocrinology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hülya Kayserili
- Medical Genetics Department, Koc University School of Medicine (KUSOM), Istanbul, Turkey
| | - Feyza Darendeliler
- Department of Pediatric Endocrinology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
2
|
Munir A, Khan IU, Rashid A, Anwar I, Shah S, Oreshkov S, Ullah M, Khan HA, Ullah U, Ahmad A, Ansar M, Rehman AU. A novel homozygous missense variant in POC1B causes cone dystrophy in a consanguineous Pakistani family. Ophthalmic Genet 2025; 46:47-55. [PMID: 39568138 DOI: 10.1080/13816810.2024.2430700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/14/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Cone dystrophy is a heterogeneous hereditary retinal disorder with disease symptoms appearing in the late first or early second decades of life. METHODS A consanguineous Pakistani family with three affected individuals underwent detailed clinical and genetic investigation. RESULTS The proband, a 63-years old male, showed severely reduced day vision, a visual acuity of counting fingers (CF), color vision deficiency, high myopia and photophobia. Fundus images showed bilateral peripapillary atrophy, bilateral dull foveal reflex, tilted disc, tessellated fundus, and hyperfluorescence at the peripheral superior temporal arcade and leak at an early stage. OCT macula and angiography findings suggested traction near the disc in the right eye and sub-retinal fluid at the fovea in the left eye. Retinal layers were normal toward the periphery but disorganized near the disc. Full visual field tests showed bilateral central scotoma, while single visual field tests indicated bilateral generalized depression of the visual field. The proband showed normal bilateral intraocular pressure, normal choroidal vessels, and unremarkable anterior segment. Exome sequencing identified a novel homozygous missense variant (POC1B:NM_172240.3:c.1391T>C;p.L464P) in the proband. Existing evidence supported pathogenicity of the identified variant in the family. CONCLUSION In conclusion, we document a first-ever Pakistani family with POC1B-related cone dystrophy.
Collapse
Affiliation(s)
- Asad Munir
- Department of Zoology, Faculty of Biological and Health Sciences, Hazara University, Mansehra, Pakistan
| | - Inam Ullah Khan
- Department of Ophthalmology, Saidu Group of Teaching Hospital, Saidu Sharif, Pakistan
| | - Abdur Rashid
- Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile Des Aveugles, Lausanne, Switzerland
| | - Ijaz Anwar
- Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile Des Aveugles, Lausanne, Switzerland
| | - Sabawoon Shah
- Department of Medicine, Gomal Medical College, DI Khan, Pakistan
| | - Sergey Oreshkov
- Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile Des Aveugles, Lausanne, Switzerland
| | - Mukhtar Ullah
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Haider Ali Khan
- Department of Ophthalmology, Saidu Group of Teaching Hospital, Saidu Sharif, Pakistan
| | - Ubaid Ullah
- Department of Ophthalmology, Saidu Group of Teaching Hospital, Saidu Sharif, Pakistan
| | - Ashfaq Ahmad
- Department of Bioinformatics, Faculty of Natural and Computational Sciences, Hazara University, Mansehra, Pakistan
| | - Muhammad Ansar
- Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile Des Aveugles, Lausanne, Switzerland
- Advanced Molecular Genetics and Genomics Disease Research and Treatment Centre, Dow University of Health Sciences, Sindh, Pakistan
| | - Atta Ur Rehman
- Department of Zoology, Faculty of Biological and Health Sciences, Hazara University, Mansehra, Pakistan
| |
Collapse
|
3
|
Sala C, Würtz M, Atorino ES, Neuner A, Partscht P, Hoffmann T, Eustermann S, Schiebel E. An interaction network of inner centriole proteins organised by POC1A-POC1B heterodimer crosslinks ensures centriolar integrity. Nat Commun 2024; 15:9857. [PMID: 39543170 PMCID: PMC11564547 DOI: 10.1038/s41467-024-54247-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
Centriole integrity, vital for cilia formation and chromosome segregation, is crucial for human health. The inner scaffold within the centriole lumen composed of the proteins POC1B, POC5 and FAM161A is key to this integrity. Here, we provide an understanding of the function of inner scaffold proteins. We demonstrate the importance of an interaction network organised by POC1A-POC1B heterodimers within the centriole lumen, where the WD40 domain of POC1B localises close to the centriole wall, while the POC5-interacting WD40 of POC1A resides in the centriole lumen. The POC1A-POC5 interaction and POC5 tetramerization are essential for inner scaffold formation and centriole stability. The microtubule binding proteins FAM161A and MDM1 by binding to POC1A-POC1B, likely positioning the POC5 tetramer near the centriole wall. Disruption of POC1A or POC1B leads to centriole microtubule defects and deletion of both genes causes centriole disintegration. These findings provide insights into organisation and function of the inner scaffold.
Collapse
Affiliation(s)
- Cornelia Sala
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Allianz, Universität Heidelberg, Heidelberg, Germany
| | - Martin Würtz
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Allianz, Universität Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL) Heidelberg, Heidelberg, Germany
| | | | - Annett Neuner
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Allianz, Universität Heidelberg, Heidelberg, Germany
| | | | - Thomas Hoffmann
- European Molecular Biology Laboratory (EMBL) Heidelberg, Heidelberg, Germany
| | | | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Allianz, Universität Heidelberg, Heidelberg, Germany.
| |
Collapse
|
4
|
Perge K, Capel E, Senée V, Julier C, Vigouroux C, Nicolino M. Ciliopathies are responsible for short stature and insulin resistance: A systematic review of this clinical association regarding SOFT syndrome. Rev Endocr Metab Disord 2024; 25:827-838. [PMID: 39017987 PMCID: PMC11470920 DOI: 10.1007/s11154-024-09894-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
SOFT syndrome (Short stature-Onychodysplasia-Facial dysmorphism-hypoTrichosis) is a rare primordial dwarfism syndrome caused by biallelic variants in POC1A encoding a centriolar protein. To refine the phenotypic spectrum of SOFT syndrome, recently shown to include metabolic features, we conducted a systematic review of all published cases (19 studies, including 42 patients). The SOFT tetrad affected only 24 patients (57%), while all cases presented with short stature from birth (median height: -5.5SDS([-8.5]-[-2.8])/adult height: 132.5 cm(103.5-148)), which was most often disproportionate (90.5%), with relative macrocephaly. Bone involvement resulted in short hands and feet (100%), brachydactyly (92.5%), metaphyseal (92%) or epiphyseal (84%) anomalies, and/or sacrum/pelvis hypoplasia (58%). Serum IGF-I was increased (median IGF-I level: + 2 SDS ([-0.5]-[+ 3])). Recombinant human growth hormone (rhGH) therapy was stopped for absence/poor growth response (7/9 patients, 78%) and/or hyperglycemia (4/9 patients, 45%). Among 11 patients evaluated, 10 (91%) presented with central distribution of fat (73%), clinical (64%) and/or biological insulin resistance (IR) (100%, median HOMA-IR: 18), dyslipidemia (80%), and hepatic steatosis (100%). Glucose tolerance abnormalities affected 58% of patients aged over 10 years. Patients harbored biallelic missense (52.4%) or truncating (45.2%) POC1A variants. Biallelic null variants, affecting 36% of patients, were less frequently associated with the SOFT tetrad (33% vs 70% respectively, p = 0.027) as compared to other variants, without difference in the prevalence of metabolic abnormalities. POC1A should be sequenced in children with short stature, altered glucose/insulin homeostasis and/or centripetal fat distribution. In patients with SOFT syndrome, rhGH treatment is not indicated, and IR-related complications should be regularly screened and monitored.PROSPERO registration: CRD42023460876.
Collapse
Affiliation(s)
- Kevin Perge
- Pediatric Endocrinology, Diabetology and Metabolism Department, Femme Mère Enfant Hospital, Hospices Civils de Lyon, Bron, France.
- Claude Bernard University, Lyon 1, Lyon, France.
- Paris University, Institut Cochin, INSERM U1016, CNRS UMR-8104, Paris, France.
| | - Emilie Capel
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Valérie Senée
- Paris University, Institut Cochin, INSERM U1016, CNRS UMR-8104, Paris, France
| | - Cécile Julier
- Paris University, Institut Cochin, INSERM U1016, CNRS UMR-8104, Paris, France
| | - Corinne Vigouroux
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition, Paris, France
- Department of Endocrinology, Diabetology and Reproductive Endocrinology, Assistance Publique-Hôpitaux de Paris, Saint-Antoine University Hospital, National Reference Center for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
- Department of Molecular Biology and Genetics, Assistance Publique-Hôpitaux de Paris, Saint-Antoine University Hospital, Paris, France
| | - Marc Nicolino
- Pediatric Endocrinology, Diabetology and Metabolism Department, Femme Mère Enfant Hospital, Hospices Civils de Lyon, Bron, France
- Claude Bernard University, Lyon 1, Lyon, France
| |
Collapse
|
5
|
Zhou H, Zhang M, Zhang X, Du X, Cao H, Bi X. Analysis of the role of POC1A in the development and progression of hepatocellular carcinoma. Transl Cancer Res 2024; 13:5003-5020. [PMID: 39430849 PMCID: PMC11483421 DOI: 10.21037/tcr-23-2398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 07/05/2024] [Indexed: 10/22/2024]
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common and deadly cancers worldwide. POC1 centriolar protein A (POC1A) is a gene encoding a protein that plays a key role in the centrosome, and is one of the two isoforms of POC1. To date, the expression of POC1A in HCC and its potential as a biomarker and tumor therapeutic target have not been examined. This study aimed to explore the effect of POC1A on patients with HCC and its potential mechanism. Methods This study investigated the role of POC1A in the occurrence and development of HCC. It analyzed the expression of POC1A in various types of HCC patients and its effect on survival using HCC patient information from the Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), the Human Protein Atlas (HPA), and the Hepatocellular Carcinoma Cell DataBase (HCCDB). It then explored the major enrichment pathways and gene functions of POC1A in HCC using the gene set enrichment analysis (GSEA) method and examined its protein-protein interactions (PPIs). Finally, it predicted the potential transcription factors (TFs) and target microRNAs (miRNAs) of POC1A, and analyzed the single nucleotide variation (SNV) and copy number variation (CNV) mutations of POC1A and the related genes in HCC, as well as their effects on immune cells. Results The results showed that POC1A was significantly overexpressed in HCC and was significantly associated with a poor prognosis. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses indicated that POC1A was mainly involved in the regulation of cell-cycle pathways and chromosome segregation functions. POC1A showed significant interactions with NUDC and PPARG, and they both had different numbers of SNV and CNV mutations in the HCC samples. In relation to immunity, the high expression of POC1A and its reciprocal genes may play an important role in B cells and macrophages. Conclusions In general, our findings suggest that POC1A overexpression could have an important effect on the development of HCC by regulating cell-cycle pathways, and that it could serve as a novel prognostic biomarker and a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Hongrui Zhou
- College of Life Science, Liaoning University, Shenyang, China
| | - Mengxue Zhang
- College of Life Science, Liaoning University, Shenyang, China
| | - Xin Zhang
- College of Life Science, Liaoning University, Shenyang, China
| | - Xintong Du
- College of Life Science, Liaoning University, Shenyang, China
| | - Huihui Cao
- College of Life Science, Liaoning University, Shenyang, China
| | - Xiuli Bi
- College of Life Science, Liaoning University, Shenyang, China
- Key Laboratory of Chronic Disease Occurrence and Nutrition Intervention, Liaoning University, Shenyang, China
| |
Collapse
|
6
|
Ruehle MD, Li S, Agard DA, Pearson CG. Poc1 bridges basal body inner junctions to promote triplet microtubule integrity and connections. J Cell Biol 2024; 223:e202311104. [PMID: 38743010 PMCID: PMC11094743 DOI: 10.1083/jcb.202311104] [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: 11/27/2023] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Basal bodies (BBs) are conserved eukaryotic structures that organize cilia. They are comprised of nine, cylindrically arranged, triplet microtubules (TMTs) connected to each other by inter-TMT linkages which stabilize the structure. Poc1 is a conserved protein important for BB structural integrity in the face of ciliary forces transmitted to BBs. To understand how Poc1 confers BB stability, we identified the precise position of Poc1 in the Tetrahymena BB and the effect of Poc1 loss on BB structure. Poc1 binds at the TMT inner junctions, stabilizing TMTs directly. From this location, Poc1 also stabilizes inter-TMT linkages throughout the BB, including the cartwheel pinhead and the inner scaffold. The full localization of the inner scaffold protein Fam161A requires Poc1. As ciliary forces are increased, Fam161A is reduced, indicative of a force-dependent molecular remodeling of the inner scaffold. Thus, while not essential for BB assembly, Poc1 promotes BB interconnections that establish an architecture competent to resist ciliary forces.
Collapse
Affiliation(s)
- Marisa D. Ruehle
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sam Li
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - David A. Agard
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood Shores, CA, USA
| | - Chad G. Pearson
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
7
|
Li Z, Li J, Wu Z, Zhu Y, Zhuo T, Nong J, Qian J, Peng H, Dai L, Wang Y, Chen M, Zeng X. Upregulation of POC1A in lung adenocarcinoma promotes tumour progression and predicts poor prognosis. J Cell Mol Med 2024; 28:e18135. [PMID: 38429900 PMCID: PMC10907829 DOI: 10.1111/jcmm.18135] [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: 09/29/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 03/03/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is characterized by a high incidence rate and mortality. Recently, POC1 centriolar protein A (POC1A) has emerged as a potential biomarker for various cancers, contributing to cancer onset and development. However, the association between POC1A and LUAD remains unexplored. We extracted The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) data sets to analyse the differential expression of POC1A and its relationship with clinical stage. Additionally, we performed diagnostic receiver operator characteristic (ROC) curve analysis and Kaplan-Meier (KM) survival analysis to assess the diagnostic and prognostic value of POC1A in LUAD. Furthermore, we investigated the correlation between POC1A expression and immune infiltration, tumour mutation burden (TMB), immune checkpoint expression and drug sensitivity. Finally, we verified POC1A expression using real-time quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC). Cell experiments were conducted to validate the effect of POC1A expression on the proliferation, migration and invasion of lung cancer cells. POC1A exhibited overexpression in most tumour tissues, and its overexpression in LUAD was significantly correlated with late-stage presentation and poor prognosis. The high POC1A expression group showed lower levels of immune infiltration but higher levels of immune checkpoint expression and TMB. Moreover, the high POC1A expression group demonstrated sensitivity to multiple drugs. In vitro experiments confirmed that POC1A knockdown led to decreased proliferation, migration, and invasion of lung cancer cells. Our findings suggest that POC1A may contribute to tumour development by modulating the cell cycle and immune cell infiltration. It also represents a potential therapeutic target and marker for the diagnosis and prognosis of LUAD.
Collapse
Affiliation(s)
- Zi‐Hao Li
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Jia‐Yi Li
- Geriatrics Department of Endocrinology and MetabolismThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Zuo‐Tao Wu
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Yong‐Jie Zhu
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Ting Zhuo
- Department of Respiratory MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Ju‐Sen Nong
- Department of Pediatric SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Jing Qian
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Hua‐Jian Peng
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Lei Dai
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Yong‐Yong Wang
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Ming‐Wu Chen
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Xiao‐Chun Zeng
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| |
Collapse
|
8
|
Perge K, Capel E, Villanueva C, Gautheron J, Diallo S, Auclair M, Rondeau S, Morichon R, Brioude F, Jéru I, Rossi M, Nicolino M, Vigouroux C. Ciliopathy due to POC1A deficiency: clinical and metabolic features, and cellular modeling. Eur J Endocrinol 2024; 190:151-164. [PMID: 38245004 DOI: 10.1093/ejendo/lvae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/08/2024] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
OBJECTIVE SOFT syndrome (MIM#614813), denoting Short stature, Onychodysplasia, Facial dysmorphism, and hypoTrichosis, is a rare primordial dwarfism syndrome caused by biallelic variants in POC1A, encoding a centriolar protein. SOFT syndrome, characterized by severe growth failure of prenatal onset and dysmorphic features, was recently associated with insulin resistance. This study aims to further explore its endocrinological features and pathophysiological mechanisms. DESIGN/METHODS We present clinical, biochemical, and genetic features of 2 unrelated patients carrying biallelic pathogenic POC1A variants. Cellular models of the disease were generated using patients' fibroblasts and POC1A-deleted human adipose stem cells. RESULTS Both patients present with clinical features of SOFT syndrome, along with hyperinsulinemia, diabetes or glucose intolerance, hypertriglyceridemia, liver steatosis, and central fat distribution. They also display resistance to the effects of IGF-1. Cellular studies show that the lack of POC1A protein expression impairs ciliogenesis and adipocyte differentiation, induces cellular senescence, and leads to resistance to insulin and IGF-1. An altered subcellular localization of insulin receptors and, to a lesser extent, IGF1 receptors could also contribute to resistance to insulin and IGF1. CONCLUSIONS Severe growth retardation, IGF-1 resistance, and centripetal fat repartition associated with insulin resistance-related metabolic abnormalities should be considered as typical features of SOFT syndrome caused by biallelic POC1A null variants. Adipocyte dysfunction and cellular senescence likely contribute to the metabolic consequences of POC1A deficiency. SOFT syndrome should be included within the group of monogenic ciliopathies with metabolic and adipose tissue involvement, which already encompasses Bardet-Biedl and Alström syndromes.
Collapse
Affiliation(s)
- Kevin Perge
- Pediatric Endocrinology, Diabetology and Metabolism Department, Femme Mère Enfant Hospital, Hospices Civils de Lyon, Bron F69500, France
- Claude Bernard University, Lyon 1, Lyon F69100, France
| | - Emilie Capel
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, and Institute of Cardiometabolism and Nutrition, F75012 Paris, France
| | - Carine Villanueva
- Pediatric Endocrinology, Diabetology and Metabolism Department, Femme Mère Enfant Hospital, Hospices Civils de Lyon, Bron F69500, France
| | - Jérémie Gautheron
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, and Institute of Cardiometabolism and Nutrition, F75012 Paris, France
| | - Safiatou Diallo
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, and Institute of Cardiometabolism and Nutrition, F75012 Paris, France
| | - Martine Auclair
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, and Institute of Cardiometabolism and Nutrition, F75012 Paris, France
| | - Sophie Rondeau
- Department of Molecular Biology, Assistance Publique-Hôpitaux de Paris, Necker Enfants Malades Hospital, Paris F75015, France
| | - Romain Morichon
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, and Institute of Cardiometabolism and Nutrition, F75012 Paris, France
- Cytometry and Imagery platform Saint-Antoine (CISA), Inserm UMS30 Lumic, Paris F75012, France
| | - Frédéric Brioude
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, and Institute of Cardiometabolism and Nutrition, F75012 Paris, France
- Department of Molecular Biology and Genetics, Assistance Publique-Hôpitaux de Paris, Armand Trousseau University Hospital, Paris F75012, France
| | - Isabelle Jéru
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, and Institute of Cardiometabolism and Nutrition, F75012 Paris, France
- Department of Molecular Biology and Genetics, Assistance Publique-Hôpitaux de Paris, Saint-Antoine University Hospital, Paris F75012, France
| | - Massimiliamo Rossi
- Genetics Department, Referral Center for Skeletal Dysplasias, Femme Mère Enfant Hospital, Hospices Civils de Lyon, Lyon F69500, France
- UMR5292, Lyon Neuroscience Research Center, INSERM U1028, CNRS, GENDEV Team, Bron F69500, France
| | - Marc Nicolino
- Pediatric Endocrinology, Diabetology and Metabolism Department, Femme Mère Enfant Hospital, Hospices Civils de Lyon, Bron F69500, France
- Claude Bernard University, Lyon 1, Lyon F69100, France
| | - Corinne Vigouroux
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, and Institute of Cardiometabolism and Nutrition, F75012 Paris, France
- Department of Molecular Biology and Genetics, Assistance Publique-Hôpitaux de Paris, Saint-Antoine University Hospital, Paris F75012, France
- Department of Endocrinology, Diabetology and Reproductive Endocrinology, Assistance Publique-Hôpitaux de Paris, Saint-Antoine University Hospital, National Reference Center for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris F75012, France
| |
Collapse
|
9
|
Alzahem TA, AlTheeb A, Ba-Abbad R. Phenotypic and genotypic features of POC1B-associated cone dystrophy. Ophthalmic Genet 2024; 45:72-77. [PMID: 37246743 DOI: 10.1080/13816810.2023.2204361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 04/14/2023] [Indexed: 05/30/2023]
Abstract
PURPOSE Patients with cone dystrophy (CD) can present with virtually normal retinal appearance, which may delay diagnosis. This study describes the inconspicuous clinical features of POC1B-associated CD in two Saudi families. METHODS This is a retrospective case study. Clinical data analyzed included multimodal retinal imaging and electroretinography of the affected individuals. Genetic analysis was done for all probands. RESULTS Three affected males from two Saudi families with POC1B-associated CD were included. The ages at presentation ranged from 18 to 34 years. Ophthalmic examination showed decreased Snellen visual acuities (range: 20/100-20/300) and color vision bilaterally. Fundus examination showed only mild vascular attenuation. Macular optical coherence tomography showed reduced reflectivity of the external limiting membrane, ellipsoid, and interdigitation zones. Full-field electroretinography demonstrated undetectable light-adapted responses and normal dark-adapted responses in all patients. Next-generation sequencing showed one proband to be homozygous for a previously unpublished nonsense variant in POC1B (NM_172240):c.672C>G; p(Tyr224*). Whole exome sequencing for the second proband showed a novel homozygous frameshifting variant in POC1B: c.991del; p(Arg331Glufs*13). CONCLUSION We described two novel variants in POC1B and the associated subtle, yet significant retinal features. POC1B-associated CD is a rare cause of visual loss in patients with relatively normal fundus appearance. Deep phenotyping is necessary in formulating appropriate differential diagnosis.
Collapse
Affiliation(s)
- Tariq A Alzahem
- Ocular Genetics Service, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
- Ophthalmology Department, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Abdulwahab AlTheeb
- Ocular Genetics Service, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - Rola Ba-Abbad
- Ocular Genetics Service, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Subbiah A, Caswell DL, Turner K, Jaiswal A, Avidor-Reiss T. CP110 and CEP135 Localize Near the Proximal Centriolar Remnants of Mice Spermatozoa. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001083. [PMID: 38351906 PMCID: PMC10862134 DOI: 10.17912/micropub.biology.001083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Centrioles form centrosomes that organize microtubules, assist in cell structure, and nucleate cilia that provide motility and sensation. Within the sperm, the centrosome consists of two centrioles (proximal and distal centriole) and a pericentriolar material known as the striated column and capitulum. The distal centriole nucleates the flagellum. Mice spermatozoa, unlike other mammal spermatozoa (e.g., human and bovine), have no ultra-structurally recognizable centrioles, but their neck has the centriolar proteins POC1B and FAM161A, suggesting mice spermatozoa have remnant centrioles. Here, we examine whether other centriolar proteins, CP110 and CEP135, found in the human and bovine spermatozoa centrioles are also found in the mouse spermatozoa neck. CP110 is a tip protein controlling ciliogenesis, and CEP135 is a centriole-specific structural protein in the centriole base of canonical centrioles found in most cell types. Here, we report that CP110 and CEP135 were both located in the mice spermatozoa neck around the proximal centriolar remnants labeled by POC1B, increasing the number of centriolar proteins found in the mice spermatozoa neck, further supporting the hypothesis that a remnant proximal centriole is present in mice.
Collapse
|
11
|
Arslanhan MD, Cengiz-Emek S, Odabasi E, Steib E, Hamel V, Guichard P, Firat-Karalar EN. CCDC15 localizes to the centriole inner scaffold and controls centriole length and integrity. J Cell Biol 2023; 222:e202305009. [PMID: 37934472 PMCID: PMC10630097 DOI: 10.1083/jcb.202305009] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/23/2023] [Accepted: 09/23/2023] [Indexed: 11/08/2023] Open
Abstract
Centrioles are microtubule-based organelles responsible for forming centrosomes and cilia, which serve as microtubule-organizing, signaling, and motility centers. Biogenesis and maintenance of centrioles with proper number, size, and architecture are vital for their functions during development and physiology. While centriole number control has been well-studied, less is understood about their maintenance as stable structures with conserved size and architecture during cell division and ciliary motility. Here, we identified CCDC15 as a centriole protein that colocalizes with and interacts with the inner scaffold, a crucial centriolar subcompartment for centriole size control and integrity. Using ultrastructure expansion microscopy, we found that CCDC15 depletion affects centriole length and integrity, leading to defective cilium formation, maintenance, and response to Hedgehog signaling. Moreover, loss-of-function experiments showed CCDC15's role in recruiting both the inner scaffold protein POC1B and the distal SFI1/Centrin-2 complex to centrioles. Our findings reveal players and mechanisms of centriole architectural integrity and insights into diseases linked to centriolar defects.
Collapse
Affiliation(s)
- Melis D. Arslanhan
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Seyma Cengiz-Emek
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Ezgi Odabasi
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Emmanuelle Steib
- Department of Bioengineering, Imperial College London, London, UK
| | - Virginie Hamel
- Department of Molecular and Cellular Biology, Sciences III, University of Geneva, Geneva, Switzerland
| | - Paul Guichard
- Department of Molecular and Cellular Biology, Sciences III, University of Geneva, Geneva, Switzerland
| | - Elif Nur Firat-Karalar
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
- Koç University School of Medicine, Istanbul, Turkey
| |
Collapse
|
12
|
Ruehle MD, Li S, Agard DA, Pearson CG. Poc1 is a basal body inner junction protein that promotes triplet microtubule integrity and interconnections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567593. [PMID: 38014135 PMCID: PMC10680851 DOI: 10.1101/2023.11.17.567593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Basal bodies (BBs) are conserved eukaryotic structures that organize motile and primary cilia. The BB is comprised of nine, cylindrically arranged, triplet microtubules (TMTs) that are connected to each other by inter-TMT linkages which maintain BB structure. During ciliary beating, forces transmitted to the BB must be resisted to prevent BB disassembly. Poc1 is a conserved BB protein important for BBs to resist ciliary forces. To understand how Poc1 confers BB stability, we identified the precise position of Poc1 binding in the Tetrahymena BB and the effect of Poc1 loss on BB structure. Poc1 binds at the TMT inner junctions, stabilizing TMTs directly. From this location, Poc1 also stabilizes inter-TMT linkages throughout the BB, including the cartwheel pinhead and the inner scaffold. Moreover, we identify a molecular response to ciliary forces via a molecular remodeling of the inner scaffold, as determined by differences in Fam161A localization. Thus, while not essential for BB assembly, Poc1 promotes BB interconnections that establish an architecture competent to resist ciliary forces.
Collapse
Affiliation(s)
- Marisa D. Ruehle
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sam Li
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - David A. Agard
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Institute for Advanced Biological Imaging, 3400 Bridge Parkway, Redwood Shores, CA, USA
| | - Chad G. Pearson
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
13
|
Kalbfuss N, Gönczy P. Towards understanding centriole elimination. Open Biol 2023; 13:230222. [PMID: 37963546 PMCID: PMC10645514 DOI: 10.1098/rsob.230222] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/14/2023] [Indexed: 11/16/2023] Open
Abstract
Centrioles are microtubule-based structures crucial for forming flagella, cilia and centrosomes. Through these roles, centrioles are critical notably for proper cell motility, signalling and division. Recent years have advanced significantly our understanding of the mechanisms governing centriole assembly and architecture. Although centrioles are typically very stable organelles, persisting over many cell cycles, they can also be eliminated in some cases. Here, we review instances of centriole elimination in a range of species and cell types. Moreover, we discuss potential mechanisms that enable the switch from a stable organelle to a vanishing one. Further work is expected to provide novel insights into centriole elimination mechanisms in health and disease, thereby also enabling scientists to readily manipulate organelle fate.
Collapse
Affiliation(s)
- Nils Kalbfuss
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
14
|
Qi X, Yuan Q, Xia X, Li W, Cao M, Yang W. Clinical and molecular analysis of cilia-associated gene signature for prognostic prediction in glioma. J Cancer Res Clin Oncol 2023; 149:11443-11455. [PMID: 37386136 DOI: 10.1007/s00432-023-05022-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/23/2023] [Indexed: 07/01/2023]
Abstract
PURPOSE Glioma is a highly malignant and unfavorable cancer in the brain. Recent evidence highlights the vital role of cilia-related pathways as novel regulators of glioma development. However, the prognostic potential of ciliary pathways in glioma is still ambiguous. In this study, we aim to construct a gene signature using cilia-related genes to facilitate the prognostication of glioma. METHODS A multi-stage approach was employed to build the ciliary gene signature for prognostication of glioma. The strategy involved the implementation of univariate, LASSO, and stepwise multivariate Cox regression analyses based on TCGA cohort, followed by independent validation in CGGA and REMBRANDT cohort. The study further revealed molecular differences at the genomic, transcriptomic, and proteomic levels between distinct groups. RESULTS A prognostic tool utilizing a 9-gene signature based on ciliary pathways was developed to assess the clinical outcomes of glioma patients. The risk scores generated by the signature demonstrated a negative correlation with patient survival rates. The validation of the signature in an independent cohort reinforced its prognostic capabilities. In-depth analysis uncovered distinctive molecular characteristics at the genomic, transcriptomic, and protein-interactive levels in the high- and low-risk groups. Furthermore, the gene signature was able to predict the sensitivity of glioma patients to conventional chemotherapeutic drugs. CONCLUSION This study has established the utility of a ciliary gene signature as a reliable prognostic predictor of glioma patient survival. Findings not only enhance our comprehension of the intricate molecular mechanisms of cilia pathways in glioma, but also hold significant clinical implications in directing chemotherapeutic strategies.
Collapse
Affiliation(s)
- Xin Qi
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuyun Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoqiang Xia
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenhao Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Muqing Cao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wanchun Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
O'Neill RS, Sodeinde AK, Welsh FC, Fagerstrom CJ, Galletta BJ, Rusan NM. Spd-2 gene duplication reveals cell-type-specific pericentriolar material regulation. Curr Biol 2023; 33:3031-3040.e6. [PMID: 37379844 DOI: 10.1016/j.cub.2023.06.020] [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: 02/07/2022] [Revised: 04/25/2023] [Accepted: 06/07/2023] [Indexed: 06/30/2023]
Abstract
Centrosomes are multi-protein organelles that function as microtubule (MT) organizing centers (MTOCs), ensuring spindle formation and chromosome segregation during cell division.1,2,3 Centrosome structure includes core centrioles that recruit pericentriolar material (PCM) that anchors γ-tubulin to nucleate MTs.1,2 In Drosophila melanogaster, PCM organization depends on proper regulation of proteins like Spd-2, which dynamically localizes to centrosomes and is required for PCM, γ-tubulin, and MTOC activity in brain neuroblast (NB) mitosis and male spermatocyte (SC) meiosis.4,5,6,7,8 Some cells have distinct requirements for MTOC activity due to differences in characteristics like cell size9,10 or whether they are mitotic or meiotic.11,12 How centrosome proteins achieve cell-type-specific functional differences is poorly understood. Previous work identified alternative splicing13 and binding partners14 as contributors to cell-type-specific differences in centrosome function. Gene duplication, which can generate paralogs with specialized functions,15,16 is also implicated in centrosome gene evolution,17 including cell-type-specific centrosome genes.18,19 To gain insight into cell-type-specific differences in centrosome protein function and regulation, we investigated a duplication of Spd-2 in Drosophila willistoni, which has Spd-2A (ancestral) and Spd-2B (derived). We find that Spd-2A functions in NB mitosis, whereas Spd-2B functions in SC meiosis. Ectopically expressed Spd-2B accumulates and functions in mitotic NBs, but ectopically expressed Spd-2A failed to accumulate in meiotic SCs, suggesting cell-type-specific differences in translation or protein stability. We mapped this failure to accumulate and function in meiosis to the C-terminal tail domain of Spd-2A, revealing a novel regulatory mechanism that can potentially achieve differences in PCM function across cell types.
Collapse
Affiliation(s)
- Ryan S O'Neill
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Afeez K Sodeinde
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA
| | - Frances C Welsh
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Graduate Program, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA 98195, USA
| | - Carey J Fagerstrom
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian J Galletta
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nasser M Rusan
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Li D, Li S, Zhou J, Zheng L, Liu G, Ding C, Yuan X. Case Report: Identification of a rare nonsense mutation in the POC1A gene by NGS in a diabetes mellitus patient. Front Genet 2023; 14:1113314. [PMID: 37056285 PMCID: PMC10086226 DOI: 10.3389/fgene.2023.1113314] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/14/2023] [Indexed: 03/30/2023] Open
Abstract
Objective: This study aimed to investigate the clinical and molecular biology of a patient with a type of diabetes mellitus caused by a mutation in the POC1A (OMIM number: 614783) gene and explore its pathogenesis and related characteristics.Methods: The patient was interviewed about his medical history and subjected to relevant examinations. Blood DNA samples were collected from the patient and his family members (parents) for trio whole-exome sequencing. Whole-exome sequencing was performed using the IDT xGen Exome Research Panel v1.0 whole-exome capture chip and sequenced using an Illumina NovaSeq 6,000 series sequencer (PE150); the sequencing coverage of the target sequence was not less than 99%. After systematic analysis and screening of the cloud platform for accurate diagnosis of genetic diseases, which integrated molecular biology annotation, biology, genetics, and clinical feature analysis, combined with a pathogenic mutation database, normal human genome database, and clinical feature database of 4,000 known genetic diseases, hundreds of thousands of gene variants were graded using the gene data analysis algorithm, a three-element grading system, and the American Society of Medical Genetics gene variant grading system. After polymerase chain reaction testing, the target sequence was verified by Sanger sequencing using an ABI3730 sequencer, and the verification result was obtained using sequence analysis software.Results: The patient had a peculiar face, a thin body, and a body mass index of 16.0 kg/m2. His fasting connecting peptide was 10.2 ug/L, his fasting insulin was 44 mIU/L, his fasting blood glucose was 10.5 mmol/L, and his glycosylated haemoglobin was 12.5%. After hospitalisation, the patient was given 0.75 g/d metformin tablets and 15 mg/d pioglitazone dispersible tablets, and his fasting blood glucose reduced to 9.2 mmol/L. After 48 U/L insulin treatment, the patient’s fasting blood glucose was reduced to 8.5 mmol/L. Genetic screening revealed that there was a pathogenic variant at the POC1A gene locus and a cytosine-to-thymine mutation at the G81 locus, turning the Arg to a termination codon and shortening the POC1A protein from 359 amino acids (aa) to 80 aa. No mutation was detected in the patient’s parents’ POC1A gene loci.Conclusion: The patient’s diabetes was caused by a POC1A gene mutation at the G81 locus, which is rarely reported in the clinic. The specific manifestations of this mutation need to be further investigated.
Collapse
|
17
|
Batu Oto B, Ağırbaşlı D, Kılıçarslan O, Celik G, Kalayci Yigin A, Seven M, Yetik H. Pigmentary retinopathy with perivascular sparing in a SOFT syndrome patient with a novel homozygous splicing variant in POC1A gene. Ophthalmic Genet 2023; 44:70-73. [PMID: 35930384 DOI: 10.1080/13816810.2022.2103837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
PURPOSE SOFT syndrome is an extremely rare inherited dwarfism syndrome. The syndrome has four major clinical manifestations: short stature, onychodysplasia, facial dysmorphism, and hypotrichosis. Herein, we report a unique case of a SOFT syndrome with findings of pigmentary retinopathy. METHODS Case report. RESULTS A 3-year boy was referred to our clinic for ophthalmologic examination from Genetic Diseases Diagnosis Center. In ophthalmic examination, anterior segment was normal bilaterally in biomicroscopy. Fundus examination revealed bilateral yellow-white punctate retinal pigment epithelium lesions located in the midperipheral retina. Macula optical coherence tomography was bilaterally normal. Whole exome sequencing (WES) analysis revealed a homozygous intronic splice site variant (c.103 + 1 G>T) in POC1A, hemizygous intronic splice site variant (c.459-5T>A) in TBX22, and a heterozygous missense variant (c.2254 C>T) in DDR2 genes. CONCLUSION There is a limited number of reported cases with SOFT syndrome and, though retinal findings in SOFT syndrome have been reported in two cases previously, none were given in detail. According to our findings, perivascular and macula sparing midperipheral retina pigment epithelium changes could be observed in patients with SOFT syndrome.
Collapse
Affiliation(s)
- Bilge Batu Oto
- Department of Ophthalmology, Cerrahpasa Medical Faculty, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Deniz Ağırbaşlı
- Department of Medical Genetics, Cerrahpasa Medical Faculty, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Oğuzhan Kılıçarslan
- Department of Ophthalmology, Cerrahpasa Medical Faculty, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Gökhan Celik
- Department of Ophthalmology, ROP Screening, Treatment and Training Center, Zeynep Kamil Maternity and Children's Disease Training and Research Hospital, Istanbul, Turkey
| | - Aysel Kalayci Yigin
- Department of Medical Genetics, Cerrahpasa Medical Faculty, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Mehmet Seven
- Department of Medical Genetics, Cerrahpasa Medical Faculty, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Hüseyin Yetik
- Department of Ophthalmology, Cerrahpasa Medical Faculty, Istanbul University- Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
18
|
Huni KC, Cheung J, Sullivan M, Robison WT, Howard KM, Kingsley K. Chemotherapeutic Drug Resistance Associated with Differential miRNA Expression of miR-375 and miR-27 among Oral Cancer Cell Lines. Int J Mol Sci 2023; 24:ijms24021244. [PMID: 36674758 PMCID: PMC9865318 DOI: 10.3390/ijms24021244] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/01/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Recent advances have suggested that non-coding miRNAs (such as miR-21, miR-27, miR-145, miR-155, miR-365, miR-375 and miR-494) may be involved in multiple aspects of oral cancer chemotherapeutic responsiveness. This study evaluated whether these specific miRNAs are correlated with oral cancer responsiveness to chemotherapies, including Paclitaxel, Cisplatin and Fluorouracil (5FU). Commercially available and well-characterized oral squamous cell carcinoma cell lines (SCC4, SCC9, SCC15, SCC25 and CAL27) revealed differing resistance and chemosensitivity to these agents-with SCC9 and SCC25 demonstrating the most resistance to all chemotherapeutic agents. SCC9 and SCC25 were also the only cell lines that expressed miR-375, and were the only cell lines that did not express miR-27. In addition, the expression of miR-375 was associated with the upregulation of Rearranged L-myc fusion (RLF) and the downregulation of Centriolar protein B (POC1), whereas lack of miR-27 expression was associated with Nucleophosmin 1 (NPM1) expression. These data have revealed important regulatory pathways and mechanisms associated with oral cancer proliferation and resistance that must be explored in future studies of potential therapeutic interventions.
Collapse
Affiliation(s)
- Kieran Caberto Huni
- Department of Advanced Education in Orthodontic Dentistry, School of Dental Medicine, University of Nevada-Las Vegas, 1700 W. Charleston Boulevard, Las Vegas, NV 89106, USA
| | - Jacky Cheung
- Department of Clinical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1700 W. Charleston Boulevard, Las Vegas, NV 89106, USA
| | - Madeline Sullivan
- Department of Clinical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1700 W. Charleston Boulevard, Las Vegas, NV 89106, USA
| | - William Taylor Robison
- Department of Clinical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1700 W. Charleston Boulevard, Las Vegas, NV 89106, USA
| | - Katherine M. Howard
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1001 Shadow Lane, Las Vegas, NV 89106, USA
| | - Karl Kingsley
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1001 Shadow Lane, Las Vegas, NV 89106, USA
- Correspondence: ; Tel.: +1-702-774-2623
| |
Collapse
|
19
|
Sperm centriole assessment identifies male factor infertility in couples with unexplained infertility – a pilot study. Eur J Cell Biol 2022; 101:151243. [DOI: 10.1016/j.ejcb.2022.151243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 12/18/2022] Open
|
20
|
Zhao Q, Gao S, Chen X, Zhu X. POC1A, prognostic biomarker of immunosuppressive microenvironment in cancer. Aging (Albany NY) 2022; 14:5195-5210. [PMID: 35748773 PMCID: PMC9271305 DOI: 10.18632/aging.204141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022]
Abstract
POC1 centriolar protein A (POC1A) effect in pan-cancer remains uncertain. The POC1A expression in normal and tumor tissues underwent analysis in this study utilizing data from the Genotype-Tissue Expression (GTEx) project and the Cancer Genome Atlas (TCGA) database. POC1A prognostic value and clinicopathological features were assessed utilizing the TCGA cohort. The relationship between immunological cell infiltration and POC1A of TCGA samples downloaded from TIMER2 and ImmuCellAI databases were observed. The relation between POC1A and immunological checkpoints genes, microsatellite instability (MSI) as well as tumor mutation burden (TMB) was also evaluated. Additionally, gene set enrichment analysis (GSEA) was utilized for exploring POC1A potential molecular mechanism in pan-cancer. In almost all 33 tumors, POCA1 showed a high expression. In most cases, high POC1A expression was linked significantly with a poor prognosis. Additionally, Tumor immune infiltration and tumors microenvironment were correlated with the expression of POC1A. In addition, TMB and MSI, as well as immune checkpoint genes in pan-cancer, were related to POC1A expression. In GSEA analysis, POC1A is implicated in cell cycle and immune-related pathways. These results might elucidate the crucial roles of POC1A in pan-cancer as a prognostic biomarker and immunotherapy target.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Shuping Gao
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Xin Chen
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Xiyan Zhu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| |
Collapse
|
21
|
Vásquez-Limeta A, Lukasik K, Kong D, Sullenberger C, Luvsanjav D, Sahabandu N, Chari R, Loncarek J. CPAP insufficiency leads to incomplete centrioles that duplicate but fragment. J Cell Biol 2022; 221:213119. [PMID: 35404385 PMCID: PMC9007748 DOI: 10.1083/jcb.202108018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 11/22/2022] Open
Abstract
Centrioles are structures that assemble centrosomes. CPAP is critical for centrosome assembly, and its mutations are found in patients with diseases such as primary microcephaly. CPAP’s centrosomal localization, its dynamics, and the consequences of its insufficiency in human cells are poorly understood. Here we use human cells genetically engineered for fast degradation of CPAP, in combination with superresolution microscopy, to address these uncertainties. We show that three independent centrosomal CPAP populations are dynamically regulated during the cell cycle. We confirm that CPAP is critical for assembly of human centrioles, but not for recruitment of pericentriolar material on already assembled centrioles. Further, we reveal that CPAP insufficiency leads to centrioles with incomplete microtubule triplets that can convert to centrosomes, duplicate, and form mitotic spindle poles, but fragment owing to loss of cohesion between microtubule blades. These findings further our basic understanding of the role of CPAP in centrosome biogenesis and help understand how CPAP aberrations can lead to human diseases.
Collapse
Affiliation(s)
- Alejandra Vásquez-Limeta
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Kimberly Lukasik
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Dong Kong
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Catherine Sullenberger
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Delgermaa Luvsanjav
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Natalie Sahabandu
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Raj Chari
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Jadranka Loncarek
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| |
Collapse
|
22
|
Mericq V, Huang-Doran I, Al-Naqeb D, Basaure J, Castiglioni C, de Bruin C, Hendriks Y, Bertini E, Alkuraya FS, Losekoot M, Al-Rubeaan K, Semple RK, Wit JM. Biallelic POC1A variants cause syndromic severe insulin resistance with muscle cramps. Eur J Endocrinol 2022; 186:543-552. [PMID: 35234134 PMCID: PMC9010808 DOI: 10.1530/eje-21-0609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 03/01/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To describe clinical, laboratory, and genetic characteristics of three unrelated cases from Chile, Portugal, and Saudi Arabia with severe insulin resistance, SOFT syndrome, and biallelic pathogenic POC1A variants. DESIGN Observational study. METHODS Probands' phenotypes, including short stature, dysmorphism, and insulin resistance, were compared with previous reports. RESULTS Cases 1 (female) and 3 (male) were homozygous for known pathogenic POC1A variants: c.649C>T, p.(Arg217Trp) and c.241C>T, p.(Arg81*), respectively. Case 2 (male) was compound heterozygous for p.(Arg217Trp) variant and the rare missense variant c.370G>A, p.(Asp124Asn). All three cases exhibited severe insulin resistance, acanthosis nigricans, elevated serum triglycerides and decreased HDL, and fatty liver, resembling three previously reported cases. All three also reported severe muscle cramps. Aggregate analysis of the six known cases with biallelic POC1A variants and insulin resistance showed decreased birth weight and length mean (s.d.): -2.8 (0.9) and -3.7 (0.9) SDS, respectively), severe short stature mean (s.d.) height: -4.9 (1.7) SDS) and moderate microcephaly (mean occipitofrontal circumference -3.0 (range: -4.7 to -1.2)). These findings were similar to those reported for patients with SOFT syndrome without insulin resistance. Muscle biopsy in Case 3 showed features of muscle involvement secondary to a neuropathic process. CONCLUSIONS Patients with SOFT syndrome can develop severe dyslipidaemic insulin resistance, independent of the exonic position of the POC1A variant. They also can develop severe muscle cramps. After diagnosis, patients should be regularly screened for insulin resistance and muscle complaints.
Collapse
Affiliation(s)
- Veronica Mericq
- Institute of Maternal and Child Research, Faculty of Medicine, University of Chile, Santiago, Chile
- Department of Pediatrics, Clinica Las Condes, Santiago, Chile
- Correspondence should be addressed to V Mericq or R K Semple; or
| | - Isabel Huang-Doran
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Dhekra Al-Naqeb
- Department of Medicine, Medical Genetic Clinic, Sultan Bin Abdulaziz Humanitarian City, Riyadh, Saudi Arabia
| | | | | | - Christiaan de Bruin
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Yvonne Hendriks
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Monique Losekoot
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - Khalid Al-Rubeaan
- Research and Scientific Centre Director, Sultan Bin Abdulaziz Humanitarian City, Riyadh, Saudi Arabia
| | - Robert K Semple
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Correspondence should be addressed to V Mericq or R K Semple; or
| | - Jan M Wit
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
23
|
Rasmussen S, Kjaer Petersen K, Kristiansen MK, Andersen JS, Aboo C, Thomsen ME, Skjoldemose E, Jørgensen NK, Stensballe A, Arendt-Nielsen L. Gold micro-particles for knee osteoarthritis. Eur J Pain 2022; 26:811-824. [PMID: 35076138 PMCID: PMC9307026 DOI: 10.1002/ejp.1909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/29/2021] [Accepted: 01/08/2022] [Indexed: 11/22/2022]
Abstract
Background This exploratory study investigates if intra‐articular injected gold microparticles in knee osteoarthritis (KOA) reduce immunomodulatory‐based pain via proteomic changes in the synovial fluid (SF) and serum. Methods Thirty patients with moderate KOA were included. Intraarticular injections with 20 mg gold microparticles (72.000 particles, 20–40 µm in diameter) using the patient's synovial fluid (SF) as carrier were performed. Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) subscores for pain, stiffness, and function were assessed at inclusion, 8 weeks and 2 years The PainDetect questionnaire, pain pressure threshold (PPT), temporal summation (TS), and conditioned pain modulation (CPM), and pain diary were assessed at inclusion and 8 weeks. Proteome analysis was performed on SF and blood samples before and after 8 weeks of treatment. Results A decrease in WOMAC scores (pain (p = 0.0001), stiffness (p = 0.0088), activity (p = 0.0001)), PainDetect (p = 0.0002) and increase in PPT (p = 0.001) and CPM (p = 0.021) and a decrease in TS (p = 0.03) were found after 8 weeks compared to inclusion assessments. At 2 years follow‐up compared to baseline there was a decrease in WOMAC scores (pain (p = 0.0001), stiffness (p = 0.007), activity (p = 0.0001)) and PainDetect (p = 0.0001). In SF, 28 different proteins were downregulated and 11 upregulated (p < 0.05) mainly associated immune response. Similarly, 31 proteins were downregulated and 1 upregulated in serum (p < 0.05) reflecting key immune response and anatomical structure development processes. No adverse effects related to the treatment were recorded. Conclusions Gold microparticles injected intra‐articular in KOA joints may provide pain relief and an inflammatory modulatory effect based on proteome changes found in SF and serum. A randomized, controlled, double‐blind study is needed to infer a conclusion. Significance This study indicates that intra‐articular gold may provide advantages in clinical practice for managing knee osteoarthritic pain. The use of intraarticular gold can add new knowledge to the treatment of inflammation and pain.
Collapse
Affiliation(s)
- Sten Rasmussen
- Department of Clinical Medicine, Aalborg University.,Department of Orthopedic Surgery, Sport and Arthroscopy, Aalborg University Hospital
| | - Kristian Kjaer Petersen
- Department of Health Science and Technology, Aalborg University.,Center for Neuroplasticity and Pain, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | | | | | | | | | | | | | | | - Lars Arendt-Nielsen
- Department of Health Science and Technology, Aalborg University.,Center for Neuroplasticity and Pain, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark.,Department of Medical Gastroenterology, Mech-Sense, Aalborg University Hospital
| |
Collapse
|
24
|
Clinical Characteristics of POC1B-Associated Retinopathy and Assignment of Pathogenicity to Novel Deep Intronic and Non-Canonical Splice Site Variants. Int J Mol Sci 2021; 22:ijms22105396. [PMID: 34065499 PMCID: PMC8160832 DOI: 10.3390/ijms22105396] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
Mutations in POC1B are a rare cause of inherited retinal degeneration. In this study, we present a thorough phenotypic and genotypic characterization of three individuals harboring putatively pathogenic variants in the POC1B gene. All patients displayed a similar, slowly progressive retinopathy (cone dystrophy or cone-rod dystrophy) with normal funduscopy but disrupted outer retinal layers on optical coherence tomography and variable age of onset. Other symptoms were decreased visual acuity and photophobia. Whole genome sequencing revealed a novel homozygous frameshift variant in one patient. Another patient was shown to harbor a novel deep intronic variant in compound heterozygous state with a previously reported canonical splice site variant. The third patient showed a novel nonsense variant and a novel non-canonical splice site variant. We aimed to validate the effect of the deep intronic variant and the non-canonical splice site variant by means of in vitro splice assays. In addition, direct RNA analysis was performed in one patient. Splicing analysis revealed that the non-canonical splice site variant c.561-3T>C leads to exon skipping while the novel deep intronic variant c.1033-327T>A causes pseudoexon activation. Our data expand the genetic landscape of POC1B mutations and confirm the benefit of genome sequencing in combination with downstream functional validation using minigene assays for the analysis of putative splice variants. In addition, we provide clinical multimodal phenotyping of the affected individuals.
Collapse
|
25
|
Li S, Zhong Y, Yang Y, He S, He W. Further phenotypic features and two novel POC1A variants in a patient with SOFT syndrome: A case report. Mol Med Rep 2021; 24:494. [PMID: 33955509 PMCID: PMC8127052 DOI: 10.3892/mmr.2021.12133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/09/2021] [Indexed: 11/05/2022] Open
Abstract
Short stature, onychodysplasia, facial dysmorphism and hypotrichosis (SOFT) syndrome is a rare autosomal recessive disease caused by POC1 centriolar protein A (POC1A) pathogenic variants. However, knowledge of genotypic and phenotypic features of SOFT syndrome remain limited as few families have been examined; therefore, the clinical identification of SOFT syndrome remains a challenge. The aim of the present case report was to investigate the genetic cause of this syndrome in a patient with a short stature, unusual facial appearance, skeletal dysplasia and sparse body hair. Giemsa banding and exome sequencing were performed to investigate the genetic background of the family. Spiral computed tomography and magnetic resonance imaging were used for investigating further phenotypic features of the patient. Exome sequencing identified that POC1A had two compound heterozygous variants, namely c.850_851insG and c.593_605delGTGGGACGTGCAT, which, to the best of our knowledge, have not been reported elsewhere. Novel phenotypes were also identified as follows: i) Metaphyseal dysplasia was alleviated (and/or even disappeared) with age; ii) the density of the femoral neck was uneven and the hyperintensity signal of the metaphysis was stripe‑like. Thus, the present case report expands the knowledge regarding phenotypic and genotypic features of SOFT syndrome.
Collapse
Affiliation(s)
- Songting Li
- Children's Healthcare Institute, Hunan Children's Hospital, University of South China, Changsha, Hunan 410007, P.R. China
| | - Yan Zhong
- Children's Healthcare Institute, Hunan Children's Hospital, University of South China, Changsha, Hunan 410007, P.R. China
| | - Yongjia Yang
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute, Hunan Children's Hospital, University of South China, Changsha, Hunan 410007, P.R. China
| | - Siping He
- Department of Radiology, Hunan Children's Hospital, University of South China, Changsha, Hunan 410007, P.R. China
| | - Wenjuan He
- Children's Healthcare Institute, Hunan Children's Hospital, University of South China, Changsha, Hunan 410007, P.R. China
| |
Collapse
|
26
|
Peturson AMC, Noel NCL, MacDonald IM. A homozygous POC1B variant causes recessive cone-rod dystrophy. Ophthalmic Genet 2021; 42:349-353. [PMID: 33657974 DOI: 10.1080/13816810.2021.1894460] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Purpose: To report a case of initial cone dystrophy that advanced to a cone-rod dystrophy with homozygous variants in the POC1B gene.Methods: Retinal structure and visual function assessments were performed using fundoscopy, spectral-domain optical coherence tomography, full field electroretinography, semi-kinetic perimetry, and Ishihara plate testing. A DNA sample was collected and sent for diagnostic molecular genetic testing with a cone-rod dystrophy panel.Results: Clinical examination and electroretinography confirmed a clinical diagnosis of cone dystrophy. Molecular genetic testing revealed homozygous variants in POC1B (c.1355 G > A, p.(Arg452Gln)). Follow-up three years later showed progression to a cone-rod dystrophy.Conclusion: Our case describes an ophthalmological phenotype associated with a homozygous POC1B missense variant and provides clinical support for variant classification.
Collapse
Affiliation(s)
- Ann-Marie C Peturson
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Nicole C L Noel
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Ian M MacDonald
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
27
|
Majore S, Agolini E, Micale L, Pascolini G, Zuppi P, Cocciadiferro D, Morlino S, Mattiuzzo M, Valiante M, Castori M, Novelli A, Grammatico P. Clinical presentation and molecular characterization of a novel patient with variant POC1A-related syndrome. Clin Genet 2021; 99:540-546. [PMID: 33372278 DOI: 10.1111/cge.13911] [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: 04/18/2020] [Revised: 12/10/2020] [Accepted: 12/24/2020] [Indexed: 12/14/2022]
Abstract
Biallelic pathogenic variants in POC1A result in SOFT (Short-stature, Onychodysplasia, Facial-dysmorphism, and hypoTrichosis) and variant POC1A-related (vPOC1A) syndromes. The latter, nowadays described in only two unrelated subjects, is associated with a restricted spectrum of variants falling in exon 10, which is naturally skipped in a specific POC1A mRNA. The synthesis of an amount of a POC1A isoform from this transcript in individuals with vPOC1A syndrome has been believed as the likely explanation for such a genotype-phenotype correlation. Here, we illustrate the clinical and molecular findings in a woman who resulted to be compound heterozygous for a recurrent frameshift variant in exon 10 and a novel variant in exon 9 of POC1A. Phenotypic characteristics of this woman included severe hyperinsulinemic dyslipidemia, acanthosis nigricans, moderate growth restriction, and dysmorphisms. These manifestations overlap the clinical features of the two previously published individuals with vPOC1A syndrome. RT-PCR analysis on peripheral blood and subsequent sequencing of the obtained amplicons demonstrated a variety of POC1A alternative transcripts that resulted to be expressed in the proband, in the healthy mother, and in controls. We illustrate the possible consequences of the two POC1A identified variants in an attempt to explain pleiotropy in vPOC1A syndrome.
Collapse
Affiliation(s)
- Silvia Majore
- Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Emanuele Agolini
- Laboratory of Medical Genetics, Department of Laboratories, Bambino Gesù Children's Hospital, Rome, Italy
| | - Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Foggia, Italy
| | - Giulia Pascolini
- Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Paolo Zuppi
- Endocrinology Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Dario Cocciadiferro
- Laboratory of Medical Genetics, Department of Laboratories, Bambino Gesù Children's Hospital, Rome, Italy
| | - Silvia Morlino
- Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Matteo Mattiuzzo
- Laboratory of Medical Genetics, Department of Laboratories, Bambino Gesù Children's Hospital, Rome, Italy
| | - Michele Valiante
- Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Foggia, Italy
| | - Antonio Novelli
- Laboratory of Medical Genetics, Department of Laboratories, Bambino Gesù Children's Hospital, Rome, Italy
| | - Paola Grammatico
- Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| |
Collapse
|
28
|
Avidor-Reiss T, Carr A, Fishman EL. The sperm centrioles. Mol Cell Endocrinol 2020; 518:110987. [PMID: 32810575 PMCID: PMC7606549 DOI: 10.1016/j.mce.2020.110987] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022]
Abstract
Centrioles are eukaryotic subcellular structures that produce and regulate massive cytoskeleton superstructures. They form centrosomes and cilia, regulate new centriole formation, anchor cilia to the cell, and regulate cilia function. These basic centriolar functions are executed in sperm cells during their amplification from spermatogonial stem cells during their differentiation to spermatozoa, and finally, after fertilization, when the sperm fuses with the egg. However, sperm centrioles exhibit many unique characteristics not commonly observed in other cell types, including structural remodeling, centriole-flagellum transition zone migration, and cell membrane association during meiosis. Here, we discuss five roles of sperm centrioles: orchestrating early spermatogenic cell divisions, forming the spermatozoon flagella, linking the spermatozoon head and tail, controlling sperm tail beating, and organizing the cytoskeleton of the zygote post-fertilization. We present the historic discovery of the centriole as a sperm factor that initiates embryogenesis, and recent genetic studies in humans and other mammals evaluating the current evidence for the five functions of sperm centrioles. We also examine information connecting the various sperm centriole functions to distinct clinical phenotypes. The emerging picture is that centrioles are essential sperm components with remarkable functional diversity and specialization that will require extensive and in-depth future studies.
Collapse
Affiliation(s)
- Tomer Avidor-Reiss
- Department of Biological Sciences, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH, USA; Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| | - Alexa Carr
- Department of Biological Sciences, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH, USA
| | | |
Collapse
|
29
|
Lu J, Huang XY, Wang YH, Xie JW, Wang JB, Lin JX, Chen QY, Cao LL, Li P, Huang CM, Zheng CH. POC1A acts as a promising prognostic biomarker associated with high tumor immune cell infiltration in gastric cancer. Aging (Albany NY) 2020; 12:18982-19011. [PMID: 33052878 PMCID: PMC7732308 DOI: 10.18632/aging.103624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/04/2020] [Indexed: 01/24/2023]
Abstract
The effect of POC1 centriolar protein A (POC1A) on gastric cancer (GC) has not been clearly defined. In this study, POC1A expression and clinical information in patients with GC were analyzed. Multiple databases were used to investigate the genes that were co-expressed with POC1A and genes whose changes co-occurred with genetic alternations of POC1A. Moreover, the TISIDB and TIMER databases were used to analyze immune infiltration. The GSE54129 GC dataset and LASSO regression model (tumor vs. normal) were employed, and 6 significant differentially expressed genes (LAMP5, CEBPB, ARMC9, PAOX, VMP1, POC1A) were identified. POC1A was selected for its high expression in adjacent tissues, which was confirmed with IHC. High POC1A expression was related to better overall and recurrence-free survival. GO and KEGG analyses demonstrated that POC1A may regulate the cell cycle, DNA replication and cell growth. Furthermore, POC1A was found to be correlated with immune infiltration levels in GC according to the TISIDB and TIMER databases. These findings indicate that POC1A acts as a tumor suppressor in GC by regulating the cell cycle and cell growth. In addition, POC1A preferentially regulates the immune infiltration of GC via several immune genes. However, the specific mechanism requires further study.
Collapse
Affiliation(s)
- Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Xiao-Yan Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Yao-Hui Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
30
|
Nguyen QPH, Liu Z, Albulescu A, Ouyang H, Zlock L, Coyaud E, Laurent E, Finkbeiner W, Moraes TJ, Raught B, Mennella V. Comparative Super-Resolution Mapping of Basal Feet Reveals a Modular but Distinct Architecture in Primary and Motile Cilia. Dev Cell 2020; 55:209-223.e7. [PMID: 33038334 DOI: 10.1016/j.devcel.2020.09.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/18/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022]
Abstract
In situ molecular architecture analysis of organelles and protein assemblies is essential to understanding the role of individual components and their cellular function, and to engineering new molecular functionalities. Through a super-resolution-driven approach, here we characterize the organization of the ciliary basal foot, an appendage of basal bodies whose main role is to provide a point of anchoring to the microtubule cytoskeleton. Quantitative image analysis shows that the basal foot is organized into three main regions linked by elongated coiled-coil proteins, revealing a conserved modular architecture in primary and motile cilia, but showing distinct features reflecting its specialized functions. Using domain-specific BioID proximity labeling and super-resolution imaging, we identify CEP112 as a basal foot protein and other candidate components of this assembly, aiding future investigations on the role of basal foot across different cilia systems.
Collapse
Affiliation(s)
- Quynh P H Nguyen
- Biochemistry Department, University of Toronto, Toronto, ON M5S1A8, Canada; Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
| | - Zhen Liu
- Biochemistry Department, University of Toronto, Toronto, ON M5S1A8, Canada; Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
| | - Alexandra Albulescu
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
| | - Hong Ouyang
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
| | - Lorna Zlock
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Etienne Coyaud
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G1L8, Canada
| | - Estelle Laurent
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G1L8, Canada
| | - Walter Finkbeiner
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Theo J Moraes
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
| | - Brian Raught
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G1L8, Canada
| | - Vito Mennella
- Biochemistry Department, University of Toronto, Toronto, ON M5S1A8, Canada; Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; NIHR Southampton Biomedical Research Center, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
31
|
Steib E, Laporte MH, Gambarotto D, Olieric N, Zheng C, Borgers S, Olieric V, Le Guennec M, Koll F, Tassin AM, Steinmetz MO, Guichard P, Hamel V. WDR90 is a centriolar microtubule wall protein important for centriole architecture integrity. eLife 2020; 9:57205. [PMID: 32946374 PMCID: PMC7500955 DOI: 10.7554/elife.57205] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022] Open
Abstract
Centrioles are characterized by a nine-fold arrangement of microtubule triplets held together by an inner protein scaffold. These structurally robust organelles experience strenuous cellular processes such as cell division or ciliary beating while performing their function. However, the molecular mechanisms underlying the stability of microtubule triplets, as well as centriole architectural integrity remain poorly understood. Here, using ultrastructure expansion microscopy for nanoscale protein mapping, we reveal that POC16 and its human homolog WDR90 are components of the microtubule wall along the central core region of the centriole. We further found that WDR90 is an evolutionary microtubule associated protein. Finally, we demonstrate that WDR90 depletion impairs the localization of inner scaffold components, leading to centriole structural abnormalities in human cells. Altogether, this work highlights that WDR90 is an evolutionary conserved molecular player participating in centriole architecture integrity. Cells are made up of compartments called organelles that perform specific roles. A cylindrical organelle called the centriole is important for a number of cellular processes, ranging from cell division to movement and signaling. Each centriole contains nine blades made up of protein filaments called microtubules, which link together to form a cylinder. This well-known structure can be found in a variety of different species. Yet, it is unclear how centrioles are able to maintain this stable architecture whilst carrying out their various different cell roles. In early 2020, a group of researchers discovered a scaffold protein at the center of centrioles that helps keep the microtubule blades stable. Further investigation suggested that another protein called WDR90 may also help centrioles sustain their cylindrical shape. However, the exact role of this protein was poorly understood. To determine the role of WDR90, Steib et al. – including many of the researchers involved in the 2020 study – used a method called Ultrastructure Expansion Microscopy to precisely locate the WDR90 protein in centrioles. This revealed that WDR90 is located on the microtubule wall of centrioles in green algae and human cells grown in the lab. Further experiments showed that the protein binds directly to microtubules and that removing WDR90 from human cells causes centrioles to lose their scaffold proteins and develop structural defects. This investigation provides fundamental insights into the structure and stability of centrioles. It shows that single proteins are key components in supporting the structural integrity of organelles and shaping their overall architecture. Furthermore, these findings demonstrate how ultrastructure expansion microscopy can be used to determine the role of individual proteins within a complex structure.
Collapse
Affiliation(s)
- Emmanuelle Steib
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Marine H Laporte
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Davide Gambarotto
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Natacha Olieric
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Celine Zheng
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Susanne Borgers
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Vincent Olieric
- Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland
| | - Maeva Le Guennec
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - France Koll
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris Sud, Université Paris-Saclay, Gif sur Yvette, France
| | - Anne-Marie Tassin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris Sud, Université Paris-Saclay, Gif sur Yvette, France
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland.,Biozentrum, University of Basel, Basel, Switzerland
| | - Paul Guichard
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Virginie Hamel
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| |
Collapse
|
32
|
Sullenberger C, Vasquez-Limeta A, Kong D, Loncarek J. With Age Comes Maturity: Biochemical and Structural Transformation of a Human Centriole in the Making. Cells 2020; 9:cells9061429. [PMID: 32526902 PMCID: PMC7349492 DOI: 10.3390/cells9061429] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
Centrioles are microtubule-based cellular structures present in most human cells that build centrosomes and cilia. Proliferating cells have only two centrosomes and this number is stringently maintained through the temporally and spatially controlled processes of centriole assembly and segregation. The assembly of new centrioles begins in early S phase and ends in the third G1 phase from their initiation. This lengthy process of centriole assembly from their initiation to their maturation is characterized by numerous structural and still poorly understood biochemical changes, which occur in synchrony with the progression of cells through three consecutive cell cycles. As a result, proliferating cells contain three structurally, biochemically, and functionally distinct types of centrioles: procentrioles, daughter centrioles, and mother centrioles. This age difference is critical for proper centrosome and cilia function. Here we discuss the centriole assembly process as it occurs in somatic cycling human cells with a focus on the structural, biochemical, and functional characteristics of centrioles of different ages.
Collapse
|
33
|
CEP44 ensures the formation of bona fide centriole wall, a requirement for the centriole-to-centrosome conversion. Nat Commun 2020; 11:903. [PMID: 32060285 PMCID: PMC7021698 DOI: 10.1038/s41467-020-14767-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 01/30/2020] [Indexed: 12/25/2022] Open
Abstract
Centrosomes are essential organelles with functions in microtubule organization that duplicate once per cell cycle. The first step of centrosome duplication is the daughter centriole formation followed by the pericentriolar material recruitment to this centriole. This maturation step was termed centriole-to-centrosome conversion. It was proposed that CEP295-dependent recruitment of pericentriolar proteins drives centriole conversion. Here we show, based on the analysis of proteins that promote centriole biogenesis, that the developing centriole structure helps drive centriole conversion. Depletion of the luminal centriole protein CEP44 that binds to the A-microtubules and interacts with POC1B affecting centriole structure and centriole conversion, despite CEP295 binding to centrioles. Impairment of POC1B, TUBE1 or TUBD1, which disturbs integrity of centriole microtubules, also prevents centriole-to-centrosome conversion. We propose that the CEP295, CEP44, POC1B, TUBE1 and TUBD1 centriole biogenesis pathway that functions in the centriole lumen and on the cytoplasmic side is essential for the centriole-to-centrosome conversion. During cell division, centrosomes duplicate and newly formed centrioles must undergo centriole-to-centrosome conversion, but the molecular details are unclear. Here, the authors report that the centriole microtubule-triplet 9-fold structure scaffolds pericentriolar proteins and permits the conversion of centrioles to fully functional centrosomes.
Collapse
|
34
|
Le Guennec M, Klena N, Gambarotto D, Laporte MH, Tassin AM, van den Hoek H, Erdmann PS, Schaffer M, Kovacik L, Borgers S, Goldie KN, Stahlberg H, Bornens M, Azimzadeh J, Engel BD, Hamel V, Guichard P. A helical inner scaffold provides a structural basis for centriole cohesion. SCIENCE ADVANCES 2020; 6:eaaz4137. [PMID: 32110738 PMCID: PMC7021493 DOI: 10.1126/sciadv.aaz4137] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/03/2019] [Indexed: 05/10/2023]
Abstract
The ninefold radial arrangement of microtubule triplets (MTTs) is the hallmark of the centriole, a conserved organelle crucial for the formation of centrosomes and cilia. Although strong cohesion between MTTs is critical to resist forces applied by ciliary beating and the mitotic spindle, how the centriole maintains its structural integrity is not known. Using cryo-electron tomography and subtomogram averaging of centrioles from four evolutionarily distant species, we found that MTTs are bound together by a helical inner scaffold covering ~70% of the centriole length that maintains MTTs cohesion under compressive forces. Ultrastructure Expansion Microscopy (U-ExM) indicated that POC5, POC1B, FAM161A, and Centrin-2 localize to the scaffold structure along the inner wall of the centriole MTTs. Moreover, we established that these four proteins interact with each other to form a complex that binds microtubules. Together, our results provide a structural and molecular basis for centriole cohesion and geometry.
Collapse
Affiliation(s)
- Maeva Le Guennec
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Nikolai Klena
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Davide Gambarotto
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Marine H. Laporte
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Anne-Marie Tassin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris Sud, Université Paris-Saclay, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Hugo van den Hoek
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Philipp S. Erdmann
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Miroslava Schaffer
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Lubomir Kovacik
- Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, University of Basel, Basel CH-4058, Switzerland
| | - Susanne Borgers
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
| | - Kenneth N. Goldie
- Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, University of Basel, Basel CH-4058, Switzerland
| | - Henning Stahlberg
- Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, University of Basel, Basel CH-4058, Switzerland
| | - Michel Bornens
- Institut Curie, PSL Research University, CNRS-UMR 144, 75005 Paris, France
| | - Juliette Azimzadeh
- Université de Paris, Institut Jacques Monod, CNRS UMR7592, 75013 Paris, France
| | - Benjamin D. Engel
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- Corresponding author. (B.D.E.); (V.H.); (P.G.)
| | - Virginie Hamel
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
- Corresponding author. (B.D.E.); (V.H.); (P.G.)
| | - Paul Guichard
- University of Geneva, Department of Cell Biology, Sciences III, Geneva, Switzerland
- Corresponding author. (B.D.E.); (V.H.); (P.G.)
| |
Collapse
|
35
|
Kameya S, Fujinami K, Ueno S, Hayashi T, Kuniyoshi K, Ideta R, Kikuchi S, Kubota D, Yoshitake K, Katagiri S, Sakuramoto H, Kominami T, Terasaki H, Yang L, Fujinami-Yokokawa Y, Liu X, Arno G, Pontikos N, Miyake Y, Iwata T, Tsunoda K. Phenotypical Characteristics of POC1B-Associated Retinopathy in Japanese Cohort: Cone Dystrophy With Normal Funduscopic Appearance. Invest Ophthalmol Vis Sci 2019; 60:3432-3446. [PMID: 31390656 DOI: 10.1167/iovs.19-26650] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Cone/cone-rod dystrophy is a large group of retinal disorders with both phonotypic and genetic heterogeneity. The purpose of this study was to characterize the phenotype of eight patients from seven families harboring POC1B mutations in a cohort of the Japan Eye Genetics Consortium (JEGC). Methods Whole-exome sequencing with targeted analyses identified homozygous or compound heterozygous mutations of the POC1B gene in 7 of 548 families in the JEGC database. Ophthalmologic examinations including the best-corrected visual acuity, perimetry, fundus photography, fundus autofluorescence imaging, optical coherence tomography, and full-field and multifocal electroretinography (ERGs) were performed. Results There were four men and four women whose median age at the onset of symptoms was 15.6 years (range, 6-23 years) and that at the time of examination was 40.3 years (range, 22-67 years). The best-corrected visual acuity ranged from -0.08 to 1.52 logMAR units. The funduscopic appearance was normal in all the cases except in one case with faint mottling in the fovea. Optical coherence tomography revealed an absence of the interdigitation zone and blurred ellipsoid zone in the posterior pole, but the foveal structures were preserved in three cases. The full-field photopic ERGs were reduced or extinguished with normal scotopic responses. The central responses of the multifocal ERGs were preserved in two cases. The diagnosis was either generalized cone dystrophy in five cases or cone dystrophy with foveal sparing in three cases. Conclusions Generalized or peripheral cone dystrophy with normal funduscopic appearance is the representative phenotype of POC1B-associated retinopathy in our cohort.
Collapse
Affiliation(s)
- Shuhei Kameya
- Department of Ophthalmology, Nippon Medical School Chiba Hokusoh Hospital, Chiba, Japan
| | - Kaoru Fujinami
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.,UCL Institute of Ophthalmology, London, United Kingdom.,Moorfields Eye Hospital, London, United Kingdom.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shinji Ueno
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Takaaki Hayashi
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuki Kuniyoshi
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osaka, Japan
| | | | - Sachiko Kikuchi
- Department of Ophthalmology, Nippon Medical School Chiba Hokusoh Hospital, Chiba, Japan.,Department of Ophthalmology, Chiba, Japan
| | - Daiki Kubota
- Department of Ophthalmology, Nippon Medical School Chiba Hokusoh Hospital, Chiba, Japan
| | - Kazutoshi Yoshitake
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Satoshi Katagiri
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Sakuramoto
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Taro Kominami
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Hiroko Terasaki
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Lizhu Yang
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Yu Fujinami-Yokokawa
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.,Graduate School of Health Management, Keio University, Kanagawa, Japan.,Division of Public Health, Yokokawa Clinic, Osaka, Japan
| | - Xiao Liu
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan.,Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Gavin Arno
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.,UCL Institute of Ophthalmology, London, United Kingdom.,Moorfields Eye Hospital, London, United Kingdom.,North East Thames Regional Genetics Service, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Nikolas Pontikos
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.,UCL Institute of Ophthalmology, London, United Kingdom.,Moorfields Eye Hospital, London, United Kingdom
| | | | - Takeshi Iwata
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Kazushige Tsunoda
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | | |
Collapse
|
36
|
A novel POC1A variant in an alternatively spliced exon causes classic SOFT syndrome: clinical presentation of seven patients. J Hum Genet 2019; 65:193-197. [DOI: 10.1038/s10038-019-0693-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/30/2019] [Accepted: 11/05/2019] [Indexed: 11/08/2022]
|
37
|
Jo KH, Jaiswal A, Khanal S, Fishman EL, Curry AN, Avidor-Reiss T. Poc1B and Sas-6 Function Together during the Atypical Centriole Formation in Drosophila melanogaster. Cells 2019; 8:cells8080841. [PMID: 31387336 PMCID: PMC6721650 DOI: 10.3390/cells8080841] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
Insects and mammals have atypical centrioles in their sperm. However, it is unclear how these atypical centrioles form. Drosophila melanogaster sperm has one typical centriole called the giant centriole (GC) and one atypical centriole called the proximal centriole-like structure (PCL). During early sperm development, centriole duplication factors such as Ana2 and Sas-6 are recruited to the GC base to initiate PCL formation. The centriolar protein, Poc1B, is also recruited at this initiation stage, but its precise role during PCL formation is unclear. Here, we show that Poc1B recruitment was dependent on Sas-6, that Poc1B had effects on cellular and PCL Sas-6, and that Poc1B and Sas-6 were colocalized in the PCL/centriole core. These findings suggest that Sas-6 and Poc1B interact during PCL formation. Co-overexpression of Ana2 and Sas-6 induced the formation of ectopic particles that contained endogenous Poc1 proteins and were composed of PCL-like structures. These structures were disrupted in Poc1 mutant flies, suggesting that Poc1 proteins stabilize the PCL-like structures. Lastly, Poc1B and Sas-6 co-overexpression also induced the formation of PCL-like structures, suggesting that they can function together during the formation of the PCL. Overall, our findings suggest that Poc1B and Sas-6 function together during PCL formation.
Collapse
Affiliation(s)
- Kyoung H Jo
- Department of Biological Sciences, University of Toledo, Toledo, OH 43607, USA
| | - Ankit Jaiswal
- Department of Biological Sciences, University of Toledo, Toledo, OH 43607, USA
| | - Sushil Khanal
- Department of Biological Sciences, University of Toledo, Toledo, OH 43607, USA
| | - Emily L Fishman
- Department of Biological Sciences, University of Toledo, Toledo, OH 43607, USA
| | - Alaina N Curry
- Department of Biological Sciences, University of Toledo, Toledo, OH 43607, USA
| | - Tomer Avidor-Reiss
- Department of Biological Sciences, University of Toledo, Toledo, OH 43607, USA.
| |
Collapse
|
38
|
Abstract
The centriole organelle consists of microtubules (MTs) that exhibit a striking 9-fold radial symmetry. Centrioles play fundamental roles across eukaryotes, notably in cell signaling, motility and division. In this Cell Science at a Glance article and accompanying poster, we cover the cellular life cycle of this organelle - from assembly to disappearance - focusing on human centrioles. The journey begins at the end of mitosis when centriole pairs disengage and the newly formed centrioles mature to begin a new duplication cycle. Selection of a single site of procentriole emergence through focusing of polo-like kinase 4 (PLK4) and the resulting assembly of spindle assembly abnormal protein 6 (SAS-6) into a cartwheel element are evoked next. Subsequently, we cover the recruitment of peripheral components that include the pinhead structure, MTs and the MT-connecting A-C linker. The function of centrioles in recruiting pericentriolar material (PCM) and in forming the template of the axoneme are then introduced, followed by a mention of circumstances in which centrioles form de novo or are eliminated.
Collapse
Affiliation(s)
- Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland
| | - Georgios N Hatzopoulos
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
39
|
Li S, Fernandez JJ, Marshall WF, Agard DA. Electron cryo-tomography provides insight into procentriole architecture and assembly mechanism. eLife 2019; 8:43434. [PMID: 30741631 PMCID: PMC6384029 DOI: 10.7554/elife.43434] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/10/2019] [Indexed: 01/03/2023] Open
Abstract
Centriole is an essential structure with multiple functions in cellular processes. Centriole biogenesis and homeostasis is tightly regulated. Using electron cryo-tomography (cryoET) we present the structure of procentrioles from Chlamydomonas reinhardtii. We identified a set of non-tubulin components attached to the triplet microtubule (MT), many are at the junctions of tubules likely to reinforce the triplet. We describe structure of the A-C linker that bridges neighboring triplets. The structure infers that POC1 is likely an integral component of A-C linker. Its conserved WD40 β-propeller domain provides attachment sites for other A-C linker components. The twist of A-C linker results in an iris diaphragm-like motion of the triplets in the longitudinal direction of procentriole. Finally, we identified two assembly intermediates at the growing ends of procentriole allowing us to propose a model for the procentriole assembly. Our results provide a comprehensive structural framework for understanding the molecular mechanisms underpinning procentriole biogenesis and assembly.
Collapse
Affiliation(s)
- Sam Li
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | | | - Wallace F Marshall
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - David A Agard
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
40
|
Sydor AM, Coyaud E, Rovelli C, Laurent E, Liu H, Raught B, Mennella V. PPP1R35 is a novel centrosomal protein that regulates centriole length in concert with the microcephaly protein RTTN. eLife 2018; 7:37846. [PMID: 30168418 PMCID: PMC6141234 DOI: 10.7554/elife.37846] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/21/2018] [Indexed: 01/02/2023] Open
Abstract
Centrosome structure, function, and number are finely regulated at the cellular level to ensure normal mammalian development. Here, we characterize PPP1R35 as a novel bona fide centrosomal protein and demonstrate that it is critical for centriole elongation. Using quantitative super-resolution microscopy mapping and live-cell imaging we show that PPP1R35 is a resident centrosomal protein located in the proximal lumen above the cartwheel, a region of the centriole that has eluded detailed characterization. Loss of PPP1R35 function results in decreased centrosome number and shortened centrioles that lack centriolar distal and microtubule wall associated proteins required for centriole elongation. We further demonstrate that PPP1R35 acts downstream of, and forms a complex with, RTTN, a microcephaly protein required for distal centriole elongation. Altogether, our study identifies a novel step in the centriole elongation pathway centered on PPP1R35 and elucidates downstream partners of the microcephaly protein RTTN.
Collapse
Affiliation(s)
| | - Etienne Coyaud
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Cristina Rovelli
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Estelle Laurent
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Helen Liu
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Ontario, Canada
| | - Vito Mennella
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada.,Department of Biochemistry, University of Toronto, Ontario, Canada
| |
Collapse
|
41
|
Vassilev A, Lee CY, Vassilev B, Zhu W, Ormanoglu P, Martin SE, DePamphilis ML. Identification of genes that are essential to restrict genome duplication to once per cell division. Oncotarget 2018; 7:34956-76. [PMID: 27144335 PMCID: PMC5085202 DOI: 10.18632/oncotarget.9008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/07/2016] [Indexed: 12/02/2022] Open
Abstract
Nuclear genome duplication is normally restricted to once per cell division, but aberrant events that allow excess DNA replication (EDR) promote genomic instability and aneuploidy, both of which are characteristics of cancer development. Here we provide the first comprehensive identification of genes that are essential to restrict genome duplication to once per cell division. An siRNA library of 21,584 human genes was screened for those that prevent EDR in cancer cells with undetectable chromosomal instability. Candidates were validated by testing multiple siRNAs and chemical inhibitors on both TP53+ and TP53- cells to reveal the relevance of this ubiquitous tumor suppressor to preventing EDR, and in the presence of an apoptosis inhibitor to reveal the full extent of EDR. The results revealed 42 genes that prevented either DNA re-replication or unscheduled endoreplication. All of them participate in one or more of eight cell cycle events. Seventeen of them have not been identified previously in this capacity. Remarkably, 14 of the 42 genes have been shown to prevent aneuploidy in mice. Moreover, suppressing a gene that prevents EDR increased the ability of the chemotherapeutic drug Paclitaxel to induce EDR, suggesting new opportunities for synthetic lethalities in the treatment of human cancers.
Collapse
Affiliation(s)
- Alex Vassilev
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-2753, USA
| | - Chrissie Y Lee
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-2753, USA.,Current address: NantBioscience, Culver City, CA 90232, USA
| | - Boris Vassilev
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-2753, USA
| | - Wenge Zhu
- Department of Biochemistry and Molecular Biology, George Washington University, Washington DC 20037, USA
| | - Pinar Ormanoglu
- National Center of Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Scott E Martin
- National Center of Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA.,Current Address: Genentech, Inc., South San Francisco, CA 94080, USA
| | - Melvin L DePamphilis
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-2753, USA
| |
Collapse
|
42
|
Kominami A, Ueno S, Kominami T, Nakanishi A, Ito Y, Fujinami K, Tsunoda K, Hayashi T, Kikuchi S, Kameya S, Iwata T, Terasaki H. Case of cone dystrophy with normal fundus appearance associated with biallelic POC1B variants. Ophthalmic Genet 2017; 39:255-262. [PMID: 29220607 DOI: 10.1080/13816810.2017.1408846] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Biallelic variants of POC1B were recently reported to cause autosomal recessive non-syndromic cone dystrophy. However, the number of studies supporting this is limited, and the clinical phenotypes of cone dystrophy have not been definitively determined. The purpose of this study was to report the phenotype of a case of POC1B-associated cone dystrophy. MATERIALS AND METHODS The medical chart of one case diagnosed with cone dystrophy was reviewed. RESULTS The patient was a 20-year-old Japanese man whose chief complaint was a progressive decrease in his central vision. His decimal best-corrected visual acuity was 0.2 for the right and 0.3 for the left. Fundus examinations showed no abnormalities. The photopic electroretinograms were nonrecordable, but the scotopic electroretinograms were within normal limits. Optical coherence tomography detected a blurry line in the region of the external limiting membrane and ellipsoid zone. Adaptive optics images showed sparsely distributed cone cells around the fovea. The patient was initially diagnosed with incomplete achromatopsia. Whole-exome sequence with targeted analysis identified new compound heterozygous mutations of c.G1355A (p R452Q) and c.C987A (pY329X) in the POC1B gene. The patient was then diagnosed with cone dystrophy. CONCLUSIONS The cone dystrophy associated with POC1B variants has features similar to achromatopsia, and genetic analyses is useful in discriminating these two diseases.
Collapse
Affiliation(s)
- Azusa Kominami
- a Department of Ophthalmology , Nagoya University Graduate School of Medicine , Nagoya , Japan
| | - Shinji Ueno
- a Department of Ophthalmology , Nagoya University Graduate School of Medicine , Nagoya , Japan
| | - Taro Kominami
- a Department of Ophthalmology , Nagoya University Graduate School of Medicine , Nagoya , Japan
| | - Ayami Nakanishi
- a Department of Ophthalmology , Nagoya University Graduate School of Medicine , Nagoya , Japan
| | - Yasuki Ito
- a Department of Ophthalmology , Nagoya University Graduate School of Medicine , Nagoya , Japan
| | - Kaoru Fujinami
- b National Institute of Sensor Organs, National Hospital Organization Tokyo Medical Center , Tokyo , Japan
| | - Kazushige Tsunoda
- b National Institute of Sensor Organs, National Hospital Organization Tokyo Medical Center , Tokyo , Japan
| | - Takaaki Hayashi
- c Department of Ophthalmology , The Jikei University School of Medicine , Tokyo , Japan
| | - Sachiko Kikuchi
- d Department of Ophthalmology , Nippon Medical School Chiba Hokusoh Hospital , Chiba , Japan
| | - Shuhei Kameya
- d Department of Ophthalmology , Nippon Medical School Chiba Hokusoh Hospital , Chiba , Japan
| | - Takeshi Iwata
- b National Institute of Sensor Organs, National Hospital Organization Tokyo Medical Center , Tokyo , Japan
| | - Hiroko Terasaki
- a Department of Ophthalmology , Nagoya University Graduate School of Medicine , Nagoya , Japan
| |
Collapse
|
43
|
Giorgio E, Rubino E, Bruselles A, Pizzi S, Rainero I, Duca S, Sirchia F, Pasini B, Tartaglia M, Brusco A. A syndromic extreme insulin resistance caused by biallelic POC1A mutations in exon 10. Eur J Endocrinol 2017; 177:K21-K27. [PMID: 28819016 DOI: 10.1530/eje-17-0431] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/27/2017] [Accepted: 08/17/2017] [Indexed: 11/08/2022]
Abstract
POC1A encodes a protein with a role in centriole assembly and stability, and in ciliogenesis. Biallelic loss-of-function mutations affecting POC1A cause SOFT syndrome, an ultra-rare condition characterized by short stature, onychodysplasia, facial dysmorphism and hypotrichosis. Using exome sequencing, we identified a homozygous frameshift mutation (c.1047_1048dupC; p.G337Rfs*25) in a patient presenting with short stature, facial hirsutism, alopecia, dyslipidemia and extreme insulin resistance. The truncating variant affected exon 10, which is retained in only two of the three POC1A-mature RNAs, due to alternative processing of the transcript. Clinical discrepancies with SOFT syndrome support the hypothesis that POC1A mutations affecting exon 10 are associated with a distinct condition, corroborating a previous hypothesis based on a similar case. Furthermore, this report provides an additional example of a genetic condition presenting with clinical heterogeneity due to alternative transcript processing. In conclusion, POC1A mutations in exon 10 should be taken into account in patients with extreme insulin resistance and short stature.
Collapse
Affiliation(s)
- Elisa Giorgio
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Elisa Rubino
- Department of Neuroscience 'Rita Levi Montalcini', University of Torino, Torino, Italy
- Koelliker Hospital, Torino, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Simone Pizzi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù IRCSS, Rome, Italy
| | - Innocenzo Rainero
- Department of Neuroscience 'Rita Levi Montalcini', University of Torino, Torino, Italy
| | | | - Fabio Sirchia
- Department of Medical Sciences, University of Torino, Torino, Italy
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Barbara Pasini
- Department of Medical Sciences, University of Torino, Torino, Italy
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù IRCSS, Rome, Italy
| | - Alfredo Brusco
- Department of Medical Sciences, University of Torino, Torino, Italy
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy
| |
Collapse
|
44
|
Chen HY, Wu CT, Tang CJC, Lin YN, Wang WJ, Tang TK. Human microcephaly protein RTTN interacts with STIL and is required to build full-length centrioles. Nat Commun 2017; 8:247. [PMID: 28811500 PMCID: PMC5558016 DOI: 10.1038/s41467-017-00305-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 06/21/2017] [Indexed: 11/09/2022] Open
Abstract
Mutations in many centriolar protein-encoding genes cause primary microcephaly. Using super-resolution and electron microscopy, we find that the human microcephaly protein, RTTN, is recruited to the proximal end of the procentriole at early S phase, and is located at the inner luminal walls of centrioles. Further studies demonstrate that RTTN directly interacts with STIL and acts downstream of STIL-mediated centriole assembly. CRISPR/Cas9-mediated RTTN gene knockout in p53-deficient cells induce amplification of primitive procentriole bodies that lack the distal-half centriolar proteins, POC5 and POC1B. Additional analyses show that RTTN serves as an upstream effector of CEP295, which mediates the loading of POC1B and POC5 to the distal-half centrioles. Interestingly, the naturally occurring microcephaly-associated mutant, RTTN (A578P), shows a low affinity for STIL binding and blocks centriole assembly. These findings reveal that RTTN contributes to building full-length centrioles and illuminate the molecular mechanism through which the RTTN (A578P) mutation causes primary microcephaly. Mutations in many centriolar protein-encoding genes cause primary microcephaly. Here the authors show that human microcephaly protein RTTN directly interacts with STIL and acts downstream of STIL-mediated centriole assembly, contributing to building full-length centrioles
Collapse
Affiliation(s)
- Hsin-Yi Chen
- Graduate Institution of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chien-Ting Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Chieh-Ju C Tang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Won-Jing Wang
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Tang K Tang
- Graduate Institution of Life Sciences, National Defense Medical Center, Taipei, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan. .,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
45
|
McKinley KL, Cheeseman IM. Large-Scale Analysis of CRISPR/Cas9 Cell-Cycle Knockouts Reveals the Diversity of p53-Dependent Responses to Cell-Cycle Defects. Dev Cell 2017; 40:405-420.e2. [PMID: 28216383 DOI: 10.1016/j.devcel.2017.01.012] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/23/2016] [Accepted: 01/23/2017] [Indexed: 12/20/2022]
Abstract
Defining the genes that are essential for cellular proliferation is critical for understanding organismal development and identifying high-value targets for disease therapies. However, the requirements for cell-cycle progression in human cells remain incompletely understood. To elucidate the consequences of acute and chronic elimination of cell-cycle proteins, we generated and characterized inducible CRISPR/Cas9 knockout human cell lines targeting 209 genes involved in diverse cell-cycle processes. We performed single-cell microscopic analyses to systematically establish the effects of the knockouts on subcellular architecture. To define variations in cell-cycle requirements between cultured cell lines, we generated knockouts across cell lines of diverse origins. We demonstrate that p53 modulates the phenotype of specific cell-cycle defects through distinct mechanisms, depending on the defect. This work provides a resource to broadly facilitate robust and long-term depletion of cell-cycle proteins and reveals insights into the requirements for cell-cycle progression.
Collapse
Affiliation(s)
- Kara L McKinley
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
46
|
Khire A, Jo KH, Kong D, Akhshi T, Blachon S, Cekic AR, Hynek S, Ha A, Loncarek J, Mennella V, Avidor-Reiss T. Centriole Remodeling during Spermiogenesis in Drosophila. Curr Biol 2016; 26:3183-3189. [PMID: 28094036 DOI: 10.1016/j.cub.2016.07.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/17/2016] [Accepted: 07/06/2016] [Indexed: 10/20/2022]
Abstract
The first cell of an animal (zygote) requires centrosomes that are assembled from paternally inherited centrioles and maternally inherited pericentriolar material (PCM) [1]. In some animals, sperm centrioles with typical ultrastructure are the origin of the first centrosomes in the zygote [2-4]. In other animals, however, sperm centrioles lose their proteins and are thought to be degenerated and non-functional during spermiogenesis [5, 6]. Here, we show that the two sperm centrioles (the giant centriole [GC] and the proximal centriole-like structure [PCL]) in Drosophila melanogaster are remodeled during spermiogenesis through protein enrichment and ultrastructure modification in parallel to previously described centrosomal reduction [7]. We found that the ultrastructure of the matured sperm (spermatozoa) centrioles is modified dramatically and that the PCL does not resemble a typical centriole. We also describe a new phenomenon of Poc1 enrichment of the atypical centrioles in the spermatozoa. Using various mutants, protein expression during spermiogenesis, and RNAi knockdown of paternal Poc1, we found that paternal Poc1 enrichment is essential for the formation of centrioles during spermiogenesis and for the formation of centrosomes after fertilization in the zygote. Altogether, these findings demonstrate that the sperm centrioles are remodeled both in their protein composition and in ultrastructure, yet they are functional and are essential for normal embryogenesis in Drosophila.
Collapse
Affiliation(s)
- Atul Khire
- Department of Biological Sciences, University of Toledo, 3050 W. Towerview Boulevard, Toledo, OH 43606, USA
| | - Kyoung H Jo
- Department of Biological Sciences, University of Toledo, 3050 W. Towerview Boulevard, Toledo, OH 43606, USA
| | - Dong Kong
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | - Tara Akhshi
- Department of Biochemistry, Cell Biology Program, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | | | - Anthony R Cekic
- Department of Biological Sciences, University of Toledo, 3050 W. Towerview Boulevard, Toledo, OH 43606, USA
| | - Sarah Hynek
- Department of Biological Sciences, University of Toledo, 3050 W. Towerview Boulevard, Toledo, OH 43606, USA
| | - Andrew Ha
- Department of Biological Sciences, University of Toledo, 3050 W. Towerview Boulevard, Toledo, OH 43606, USA
| | - Jadranka Loncarek
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | - Vito Mennella
- Department of Biochemistry, Cell Biology Program, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Tomer Avidor-Reiss
- Department of Biological Sciences, University of Toledo, 3050 W. Towerview Boulevard, Toledo, OH 43606, USA.
| |
Collapse
|
47
|
Pedersen MT, Kooistra SM, Radzisheuskaya A, Laugesen A, Johansen JV, Hayward DG, Nilsson J, Agger K, Helin K. Continual removal of H3K9 promoter methylation by Jmjd2 demethylases is vital for ESC self-renewal and early development. EMBO J 2016; 35:1550-64. [PMID: 27266524 DOI: 10.15252/embj.201593317] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/06/2016] [Indexed: 12/12/2022] Open
Abstract
Chromatin-associated proteins are essential for the specification and maintenance of cell identity. They exert these functions through modulating and maintaining transcriptional patterns. To elucidate the functions of the Jmjd2 family of H3K9/H3K36 histone demethylases, we generated conditional Jmjd2a/Kdm4a, Jmjd2b/Kdm4b and Jmjd2c/Kdm4c/Gasc1 single, double and triple knockout mouse embryonic stem cells (ESCs). We report that while individual Jmjd2 family members are dispensable for ESC maintenance and embryogenesis, combined deficiency for specifically Jmjd2a and Jmjd2c leads to early embryonic lethality and impaired ESC self-renewal, with spontaneous differentiation towards primitive endoderm under permissive culture conditions. We further show that Jmjd2a and Jmjd2c both localize to H3K4me3-positive promoters, where they have widespread and redundant roles in preventing accumulation of H3K9me3 and H3K36me3. Jmjd2 catalytic activity is required for ESC maintenance, and increased H3K9me3 levels in knockout ESCs compromise the expression of several Jmjd2a/c targets, including genes that are important for ESC self-renewal. Thus, continual removal of H3K9 promoter methylation by Jmjd2 demethylases represents a novel mechanism ensuring transcriptional competence and stability of the pluripotent cell identity.
Collapse
Affiliation(s)
- Marianne Terndrup Pedersen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Marije Kooistra
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Aliaksandra Radzisheuskaya
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Anne Laugesen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Vilstrup Johansen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Daniel Geoffrey Hayward
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Nilsson
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karl Agger
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Helin
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Meehl JB, Bayless BA, Giddings TH, Pearson CG, Winey M. Tetrahymena Poc1 ensures proper intertriplet microtubule linkages to maintain basal body integrity. Mol Biol Cell 2016; 27:2394-403. [PMID: 27251062 PMCID: PMC4966981 DOI: 10.1091/mbc.e16-03-0165] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/27/2016] [Indexed: 12/31/2022] Open
Abstract
Basal bodies comprise nine symmetric triplet microtubules that anchor forces produced by the asymmetric beat pattern of motile cilia. The ciliopathy protein Poc1 stabilizes basal bodies through an unknown mechanism. In poc1∆ cells, electron tomography reveals subtle defects in the organization of intertriplet linkers (A-C linkers) that connect adjacent triplet microtubules. Complete triplet microtubules are lost preferentially near the posterior face of the basal body. Basal bodies that are missing triplets likely remain competent to assemble new basal bodies with nine triplet microtubules, suggesting that the mother basal body microtubule structure does not template the daughter. Our data indicate that Poc1 stabilizes basal body triplet microtubules through linkers between neighboring triplets. Without this stabilization, specific triplet microtubules within the basal body are more susceptible to loss, probably due to force distribution within the basal body during ciliary beating. This work provides insights into how the ciliopathy protein Poc1 maintains basal body integrity.
Collapse
Affiliation(s)
- Janet B Meehl
- Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309
| | - Brian A Bayless
- Department of Cell and Developmental Biology, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045
| | - Thomas H Giddings
- Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309
| | - Chad G Pearson
- Department of Cell and Developmental Biology, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045
| | - Mark Winey
- Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309
| |
Collapse
|
49
|
Geister KA, Brinkmeier ML, Cheung LY, Wendt J, Oatley MJ, Burgess DL, Kozloff KM, Cavalcoli JD, Oatley JM, Camper SA. LINE-1 Mediated Insertion into Poc1a (Protein of Centriole 1 A) Causes Growth Insufficiency and Male Infertility in Mice. PLoS Genet 2015; 11:e1005569. [PMID: 26496357 PMCID: PMC4619696 DOI: 10.1371/journal.pgen.1005569] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 09/15/2015] [Indexed: 11/19/2022] Open
Abstract
Skeletal dysplasias are a common, genetically heterogeneous cause of short stature that can result from disruptions in many cellular processes. We report the identification of the lesion responsible for skeletal dysplasia and male infertility in the spontaneous, recessive mouse mutant chagun. We determined that Poc1a, encoding protein of the centriole 1a, is disrupted by the insertion of a processed Cenpw cDNA, which is flanked by target site duplications, suggestive of a LINE-1 retrotransposon-mediated event. Mutant fibroblasts have impaired cilia formation and multipolar spindles. Male infertility is caused by defective spermatogenesis early in meiosis and progressive germ cell loss. Spermatogonial stem cell transplantation studies revealed that Poc1a is essential for normal function of both Sertoli cells and germ cells. The proliferative zone of the growth plate is small and disorganized because chondrocytes fail to re-align after cell division and undergo increased apoptosis. Poc1a and several other genes associated with centrosome function can affect the skeleton and lead to skeletal dysplasias and primordial dwarfisms. This mouse mutant reveals how centrosome dysfunction contributes to defects in skeletal growth and male infertility.
Collapse
Affiliation(s)
- Krista A. Geister
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Michelle L. Brinkmeier
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Leonard Y. Cheung
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jennifer Wendt
- Roche NimbleGen, Inc., Research and Development, Madison, Wisconsin, United States of America
| | - Melissa J. Oatley
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington, United States of America
| | - Daniel L. Burgess
- Roche NimbleGen, Inc., Research and Development, Madison, Wisconsin, United States of America
| | - Kenneth M. Kozloff
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| | - James D. Cavalcoli
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jon M. Oatley
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington, United States of America
| | - Sally A. Camper
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
50
|
Chen JH, Segni M, Payne F, Huang-Doran I, Sleigh A, Adams C, Savage DB, O'Rahilly S, Semple RK, Barroso I. Truncation of POC1A associated with short stature and extreme insulin resistance. J Mol Endocrinol 2015; 55:147-58. [PMID: 26336158 PMCID: PMC4722288 DOI: 10.1530/jme-15-0090] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We describe a female proband with primordial dwarfism, skeletal dysplasia, facial dysmorphism, extreme dyslipidaemic insulin resistance and fatty liver associated with a novel homozygous frameshift mutation in POC1A, predicted to affect two of the three protein products of the gene. POC1A encodes a protein associated with centrioles throughout the cell cycle and implicated in both mitotic spindle and primary ciliary function. Three homozygous mutations affecting all isoforms of POC1A have recently been implicated in a similar syndrome of primordial dwarfism, although no detailed metabolic phenotypes were described. Primary cells from the proband we describe exhibited increased centrosome amplification and multipolar spindle formation during mitosis, but showed normal DNA content, arguing against mitotic skipping, cleavage failure or cell fusion. Despite evidence of increased DNA damage in cells with supernumerary centrosomes, no aneuploidy was detected. Extensive centrosome clustering both at mitotic spindles and in primary cilia mitigated the consequences of centrosome amplification, and primary ciliary formation was normal. Although further metabolic studies of patients with POC1A mutations are warranted, we suggest that POC1A may be added to ALMS1 and PCNT as examples of centrosomal or pericentriolar proteins whose dysfunction leads to extreme dyslipidaemic insulin resistance. Further investigation of links between these molecular defects and adipose tissue dysfunction is likely to yield insights into mechanisms of adipose tissue maintenance and regeneration that are critical to metabolic health.
Collapse
Affiliation(s)
- Jian-Hua Chen
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| | - Maria Segni
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| | - Felicity Payne
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| | - Isabel Huang-Doran
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| | - Alison Sleigh
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| | - Claire Adams
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| | | | - David B Savage
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| | - Stephen O'Rahilly
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| | - Robert K Semple
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| | - Inês Barroso
- The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK The University of Cambridge Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK The National Institute for Health Research Cambridge Biomedical Research Centre Cambridge, UK Department of Pediatrics Sapienza University, Rome, Italy Metabolic Disease Group Wellcome Trust Sanger Institute, Cambridge, UK Wolfson Brain Imaging Centre University of Cambridge, Cambridge, UK National Institute for Health Research/Wellcome Trust Clinical Research Facility Cambridge, UK
| |
Collapse
|