1
|
Khan NT, Hasan Akash MM, Sajib AA, Akhteruzzaman S. Allele-specific detection of isoniazid metabolism modulating variants of N-acetyltransferase 2 enzyme and their frequencies in the Bangladeshi population. Gene 2025; 957:149480. [PMID: 40204038 DOI: 10.1016/j.gene.2025.149480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Tuberculosis is one of the oldest diseases that still affects millions of people worldwide and remains a significant public health challenge. The N-acetyltransferase 2 (NAT2) enzyme metabolizes isoniazid (INH), a primary antibiotic in tuberculosis treatment. The single nucleotide polymorphisms (SNPs) of NAT2 affect the metabolism and function of isoniazid. The rs1801280 (T341C) and rs1208 (G803A) variants of NAT2 are associated with INH drug responses. Individuals with the slow-metabolizing rs1801280 variant of the NAT2 enzyme are at a higher risk of INH-induced liver damage and require lower doses or longer treatment regimens. At the same time, individuals with the fast-metabolizing rs1208 variant are at risk of treatment failure due to rapid drug metabolism. Genotyping of the NAT2 variants can help clinicians personalize tuberculosis treatment, optimize drug doses, and thus minimize adverse effects. Under this study, an allele-specific PCR (ASPCR) method was developed for genotyping the NAT2 variants, and the results were validated through targeted sequencing. The allele frequencies at the rs1801280 locus were 0.60 for the T allele and 0.40 for the C. For rs1208, the participants' allele frequencies were 0.27 for the G allele and 0.73 for the A allele. This ASPCR method is quick, affordable, and could be used in routine genotyping to personalize the treatment for tuberculosis patients, leading to more effective and safer treatments. We also used molecular docking to study how the rs1801280 and rs1208 variants affect the interaction between the NAT2 enzyme and drugs. A slight change was visible in the flexibility of the amino acid residues. However, those amino acids were not involved in the ligand binding mechanism.
Collapse
Affiliation(s)
- Nabiha Tasneem Khan
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh; Biotechnology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | | | - Abu Ashfaqur Sajib
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Sharif Akhteruzzaman
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh.
| |
Collapse
|
2
|
Kmetsch LM, Tietze H, Bogner E. Identification of a binding pocket of letermovir in the terminase subunit pUL56 of human cytomegalovirus. Sci Rep 2025; 15:10334. [PMID: 40133489 PMCID: PMC11937570 DOI: 10.1038/s41598-025-94809-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
A key step in replication of human cytomegalovirus (HCMV) is the generation and packaging of unit-length genomes into preformed capsids. The enzymes involved in this process are viral terminases. The HCMV terminase consists of two subunits, the ATPase pUL56 and the nuclease pUL89. A potential third component, pUL51, has been proposed. Letermovir is the first terminase inhibitor available for HCMV prophylaxis to allogenic hematopoietic stem cell recipients. However, mutations in the HCMV terminase subunit pUL56 and, to a lesser extent, in pUL89 or pUL51 lead to resistance. Here we focused on the drug target area in the terminase subunit pUL56. To gain further structural insights into the putative binding site of letermovir, in silico analysis of the structure was performed using Phyre2 and SwissDock. For our analysis, we used three of the most frequent mutations during letermovir treatment, C325F, C325Y and C325W. We demonstrated that all variants have a pronounced cavity reduction, leading to the letermovir binding conformations being "pushed-out" of the binding pocket. This results in a changed distribution of the Gibbs free binding energy. To circumvent the absolute resistance of C325 mutations a further modification of letermovir might solve the problem and leads to optimizing drug targeting capacity.
Collapse
Affiliation(s)
- Lukas M Kmetsch
- Institute of Virology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, 10117, Berlin, Germany
| | - Hans Tietze
- Institute of Virology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, 10117, Berlin, Germany
| | - Elke Bogner
- Institute of Virology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, 10117, Berlin, Germany.
| |
Collapse
|
3
|
Sharma D, Singh H, Arya A, Choudhary H, Guleria P, Saini S, Thakur CJ. Comprehensive computational analysis of deleterious nsSNPs in PTEN gene for structural and functional insights. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2025; 14:219-239. [PMID: 40321699 PMCID: PMC12046362 DOI: 10.22099/mbrc.2025.52148.2092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Single nucleotide polymorphisms (SNPs) are pivotal in understanding the genetic basis of complex disorders. Among them, nonsynonymous SNPs (nsSNPs) that alter amino acid sequences can significantly impact protein structure and function. This study focuses on analyzing deleterious nsSNPs in the tumor suppressor gene PTEN (Phosphatase and TENsin Homolog), which plays a central role in regulating the PI3K/Akt signaling pathway and tumorigenesis. Out of 43,855 SNPs in PTEN, 17 deleterious nsSNPs were identified using six computational tools. Protein stability analysis revealed that 15 variants reduce stability, potentially leading to functional impairment. Structural evaluations using HOPE and ConSurf classified mutations into buried structural residues disrupting protein integrity and exposed functional residues affecting molecular interactions. STRING database analysis highlighted PTEN as a central node in an intricate protein network, with deleterious mutations impairing critical interactions with partners such as PIK3CA, AKT1, and TP53. Secondary structure analysis revealed distinct structural deviations, particularly for G129E, which exhibited the most pronounced destabilization. Molecular dynamics simulations confirmed stability variations across mutants, with G129E exhibiting greater instability. This comprehensive analysis enhances understanding of PTEN nsSNP impacts, offering insights for therapeutic interventions and future experimental validation.
Collapse
Affiliation(s)
- Divyanshi Sharma
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32C, 160030, Chandigarh, India
| | - Harasees Singh
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32C, 160030, Chandigarh, India
| | - Aryan Arya
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32C, 160030, Chandigarh, India
| | - Himanshi Choudhary
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32C, 160030, Chandigarh, India
| | - Pragya Guleria
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32C, 160030, Chandigarh, India
| | - Sandeep Saini
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32C, 160030, Chandigarh, India
- Department of Biophysics, Panjab University, Sector 25, 160014, Chandigarh, India
| | - Chander Jyoti Thakur
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32C, 160030, Chandigarh, India
| |
Collapse
|
4
|
Hasan M, Sarker MN, Jabin T, Sarker S, Ahmed S, Abdullah-Al-Shoeb M, Hossain T. Pathogenic single nucleotide polymorphisms in RhoA gene: Insights into structural and functional impacts on RhoA-PLD1 interaction through molecular dynamics simulation. Curr Res Struct Biol 2024; 8:100159. [PMID: 39698059 PMCID: PMC11653153 DOI: 10.1016/j.crstbi.2024.100159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/03/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
Molecular switches serve as key regulators of biological systems by acting as one of the crucial driving forces in the initiation of signal transduction pathway cascades. The Ras homolog gene family member A (RhoA) is one of the molecular switches that binds with GTP in order to cycle between an active GTP-bound state and an inactive GDP-bound state. Any aberrance in control over this circuit, particularly due to any perturbation in switching, leads to the development of different pathogenicity. Consequently, the single nucleotide polymorphisms (SNPs) within the RhoA gene, especially deleterious genetic variations, are crucial to study to forecast structural alteration and their functional impacts in light of disease onset. In this comprehensive study, we employed a range of computational tools to screen the deleterious SNPs of RhoA from 207 nonsynonymous SNPs (nsSNPs). By utilizing 7 distinct tools for further analysis, 8 common deleterious SNPs were sorted, among them 5 nsSNPs (V9G, G17E, E40K, A61T, F171L) were found to be in the highly conserved regions, with E40K and A61T at G2 and G3 motif of the GTP-binding domain respectively, indicating potential perturbation in GTP/GDP binding ability of the protein. RhoA-GDP complex interacts with the enzyme phospholipase, specifically PLD1, to regulate different cellular activities. PLD1 is also a crucial regulator of thrombosis and cancer. In that line of focus, our initial structural analysis of Y66H, A61T, G17E, I86N, and I151T mutations of RhoA revealed remarkable decreased hydrophobicity from which we further filtered out G17E and I86N which may have potential impact on the RhoA-GDP-PLD1 complex. Intriguingly, the comparative 250 ns (ns) molecular dynamics (MD) simulation of these two mutated complexes revealed overall structural instability and altered interaction patterns. Therefore, further investigation into these deleterious mutations with in vitro and in vivo studies could lead to the identification of potential biomarkers in terms of different pathogenesis and could also be utilized in personalized therapeutic targets in the long run.
Collapse
Affiliation(s)
| | | | | | - Saifuddin Sarker
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Shamim Ahmed
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mohammad Abdullah-Al-Shoeb
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Tanvir Hossain
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| |
Collapse
|
5
|
Ransmayr B, Bal SK, Thian M, Svaton M, van de Wetering C, Hafemeister C, Segarra-Roca A, Block J, Frohne A, Krolo A, Altunbas MY, Bilgic-Eltan S, Kıykım A, Aydiner O, Kesim S, Inanir S, Karakoc-Aydiner E, Ozen A, Aba Ü, Çomak A, Tuğcu GD, Pazdzior R, Huber B, Farlik M, Kubicek S, von Bernuth H, Simonitsch-Klupp I, Rizzi M, Halbritter F, Tumanov AV, Kraakman MJ, Metin A, Castanon I, Erman B, Baris S, Boztug K. LTβR deficiency causes lymph node aplasia and impaired B cell differentiation. Sci Immunol 2024; 9:eadq8796. [PMID: 39576873 DOI: 10.1126/sciimmunol.adq8796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024]
Abstract
Secondary lymphoid organs (SLOs) provide the confined microenvironment required for stromal cells to interact with immune cells to initiate adaptive immune responses resulting in B cell differentiation. Here, we studied three patients from two families with functional hyposplenism, absence of tonsils, and complete lymph node aplasia, leading to recurrent bacterial and viral infections. We identified biallelic loss-of-function mutations in LTBR, encoding the lymphotoxin beta receptor (LTβR), primarily expressed on stromal cells. Patients with LTβR deficiency had hypogammaglobulinemia, diminished memory B cells, regulatory and follicular T helper cells, and dysregulated expression of several tumor necrosis factor family members. B cell differentiation in an ex vivo coculture system was intact, implying that the observed B cell defects were not intrinsic in nature and instead resulted from LTβR-dependent stromal cell interaction signaling critical for SLO formation. Collectively, we define a human inborn error of immunity caused primarily by a stromal defect affecting the development and function of SLOs.
Collapse
Affiliation(s)
- Bernhard Ransmayr
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Sevgi Köstel Bal
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Marini Thian
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Michael Svaton
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Cheryl van de Wetering
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | | | - Jana Block
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | | | - Ana Krolo
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Melek Yorgun Altunbas
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University School of Medicine, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, Istanbul, Turkey
- Işıl Berat Barlan Center for Translational Medicine, Marmara University, Istanbul, Turkey
- Marmara University, Immune Deficiency Application and Research Center, Istanbul, Turkey
| | - Sevgi Bilgic-Eltan
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University School of Medicine, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, Istanbul, Turkey
- Işıl Berat Barlan Center for Translational Medicine, Marmara University, Istanbul, Turkey
- Marmara University, Immune Deficiency Application and Research Center, Istanbul, Turkey
| | - Ayça Kıykım
- Istanbul University-Cerrahpasa, Faculty of Medicine, Department of Pediatric Allergy and Immunology, Istanbul, Turkey
| | - Omer Aydiner
- Kartal Dr. Lütfi Kırdar City Hospital, Department of Radiology, Istanbul, Turkey
| | - Selin Kesim
- Marmara University, Faculty of Medicine, Department of Nuclear Medicine, Istanbul, Turkey
| | - Sabahat Inanir
- Marmara University, Faculty of Medicine, Department of Nuclear Medicine, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University School of Medicine, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, Istanbul, Turkey
- Işıl Berat Barlan Center for Translational Medicine, Marmara University, Istanbul, Turkey
- Marmara University, Immune Deficiency Application and Research Center, Istanbul, Turkey
| | - Ahmet Ozen
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University School of Medicine, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, Istanbul, Turkey
- Işıl Berat Barlan Center for Translational Medicine, Marmara University, Istanbul, Turkey
- Marmara University, Immune Deficiency Application and Research Center, Istanbul, Turkey
| | - Ümran Aba
- Can Sucak Research Laboratory for Translational Immunology, Hacettepe University, Ankara, Turkey
- Institute of Child Health, Hacettepe University, Ankara, Turkey
| | - Aylin Çomak
- Ankara Bilkent City Hospital, Children's Hospital, Department of Nuclear Medicine, Ankara, Turkey
| | - Gökçen Dilşa Tuğcu
- Ankara Bilkent City Hospital, Children's Hospital, Department of Pediatric Pulmonology, Ankara, Turkey
| | - Robert Pazdzior
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bettina Huber
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Matthias Farlik
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Horst von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité University Medicine, Berlin, Corporate Member of Free University and Humboldt University and Berlin Institute of Health, Berlin, Germany
- Labor Berlin Charité-Vivantes, Department of Immunology, Berlin, Germany
- Berlin Institute of International Health Global Health Center Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| | | | - Marta Rizzi
- Department of Rheumatology and Clinical Immunology and Center for Chronic Immunodeficiency, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | | | - Alexei V Tumanov
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michael J Kraakman
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Ayşe Metin
- Ankara Bilkent City Hospital, Children's Hospital, Department of Pediatric Immunology and Allergy, Ankara, Turkey
| | - Irinka Castanon
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Baran Erman
- Can Sucak Research Laboratory for Translational Immunology, Hacettepe University, Ankara, Turkey
- Institute of Child Health, Hacettepe University, Ankara, Turkey
| | - Safa Baris
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University School of Medicine, Istanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, Istanbul, Turkey
- Işıl Berat Barlan Center for Translational Medicine, Marmara University, Istanbul, Turkey
- Marmara University, Immune Deficiency Application and Research Center, Istanbul, Turkey
| | - Kaan Boztug
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- Medical University of Vienna, Department of Pediatrics and Adolescent Medicine, Vienna, Austria
- St. Anna Children's Hospital, Vienna, Austria
| |
Collapse
|
6
|
Rasool M, Pushparaj PN, Haque A, Shorbaji AM, Mira LS, Bakhashab S, Alama MN, Farooq M, Karim S, Larsen LA. Discovery of a novel mutation F184S (c.551T>C) in GATA4 gene causing congenital heart disease in a consanguineous Saudi family. Heliyon 2024; 10:e37177. [PMID: 39286212 PMCID: PMC11403501 DOI: 10.1016/j.heliyon.2024.e37177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Background & aim Congenital heart disease (CHD) is the most common cause of non-infectious deaths in infants worldwide. However, the molecular mechanisms underlying CHD remain unclear. Approximately 30 % of the causes are believed to be genetic mutations and chromosomal abnormalities. In this study, we aimed to identify the genetic causes of CHD in consanguineous families. Methods Fourth-generation pedigrees with CHD were recruited. The main cardiac features of the patient included absence of the right pulmonary artery and a large dilated left pulmonary artery. To determine the underlying genetic cause, whole-exome sequencing was performed and subsequently confirmed using Sanger sequencing and different online databases to study the pathogenesis of the identified gene mutation. An in-silico homology model was created using the Alphafold homology model structure of GATA4 (AF-P43694-F1). The missense3D online program was used to evaluate the structural alterations. Results We identified a deleterious mutation c.551T > C (p.Phe184Ser) in GATA4. GATA4 is a highly conserved zinc-finger transcription factor, and its continuous expression is essential for cardiogenesis during embryogenesis. The in-silico model suggested a compromised binding efficiency with other proteins. Several variant interpretation algorithms indicated that the F184S missense variant in GATA4 is damaging, whereas HOPE analysis indicated the functional impairment of DNA binding of transcription factors and zinc-ion binding activities of GATA4. Conclusion The variant identified in GATA4 appears to cause recessive CHD in the family. In silico analysis suggested that this variant was damaging and caused multiple structural and functional aberrations. This study may support prenatal screening of the fetus in this family to prevent diseases in new generations.
Collapse
Affiliation(s)
- Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Absarul Haque
- King Fahd Medical Research Center, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayat Mohammed Shorbaji
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Loubna Siraj Mira
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherin Bakhashab
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed Nabil Alama
- Department of Cardiology, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Muhammad Farooq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Lars Allan Larsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| |
Collapse
|
7
|
Longobardi A, Bellini S, Nicsanu R, Pilotto A, Geviti A, Facconi A, Tolassi C, Libri I, Saraceno C, Fostinelli S, Borroni B, Padovani A, Binetti G, Ghidoni R. Unveiling New Genetic Variants Associated with Age at Onset in Alzheimer's Disease and Frontotemporal Lobar Degeneration Due to C9orf72 Repeat Expansions. Int J Mol Sci 2024; 25:7457. [PMID: 39000564 PMCID: PMC11242823 DOI: 10.3390/ijms25137457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Alzheimer's disease (AD) and Frontotemporal lobar degeneration (FTLD) represent the most common forms of neurodegenerative dementias with a highly phenotypic variability. Herein, we investigated the role of genetic variants related to the immune system and inflammation as genetic modulators in AD and related dementias. In patients with sporadic AD/FTLD (n = 300) and GRN/C9orf72 mutation carriers (n = 80), we performed a targeted sequencing of 50 genes belonging to the immune system and inflammation, selected based on their high expression in brain regions and low tolerance to genetic variation. The linear regression analyses revealed two genetic variants: (i) the rs1049296 in the transferrin (TF) gene, shown to be significantly associated with age at onset in the sporadic AD group, anticipating the disease onset of 4 years for each SNP allele with respect to the wild-type allele, and (ii) the rs7550295 in the calsyntenin-1 (CLSTN1) gene, which was significantly associated with age at onset in the C9orf72 group, delaying the disease onset of 17 years in patients carrying the SNP allele. In conclusion, our data support the role of genetic variants in iron metabolism (TF) and in the modulation of the calcium signalling/axonal anterograde transport of vesicles (CLSTN1) as genetic modulators in AD and FTLD due to C9orf72 expansions.
Collapse
Affiliation(s)
- Antonio Longobardi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| | - Sonia Bellini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| | - Roland Nicsanu
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Hospital, 25123 Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili Hospital, 25123 Brescia, Italy
| | - Andrea Geviti
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.G.); (A.F.)
| | - Alessandro Facconi
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.G.); (A.F.)
| | - Chiara Tolassi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Hospital, 25123 Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili Hospital, 25123 Brescia, Italy
| | - Ilenia Libri
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
| | - Claudia Saraceno
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| | - Silvia Fostinelli
- MAC-Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy;
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
- Cognitive and Behavioural Neurology, ASST Spedali Civili Hospital, 25123 Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Hospital, 25123 Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili Hospital, 25123 Brescia, Italy
- Brain Health Center, University of Brescia, 25123 Brescia, Italy
| | - Giuliano Binetti
- MAC-Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy;
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| |
Collapse
|
8
|
Stephenson JD, Totoo P, Burke D, Jänes J, Beltrao P, Martin M. ProtVar: mapping and contextualizing human missense variation. Nucleic Acids Res 2024; 52:W140-W147. [PMID: 38769064 PMCID: PMC11223857 DOI: 10.1093/nar/gkae413] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/22/2024] Open
Abstract
Genomic variation can impact normal biological function in complex ways and so understanding variant effects requires a broad range of data to be coherently assimilated. Whilst the volume of human variant data and relevant annotations has increased, the corresponding increase in the breadth of participating fields, standards and versioning mean that moving between genomic, coding, protein and structure positions is increasingly complex. In turn this makes investigating variants in diverse formats and assimilating annotations from different resources challenging. ProtVar addresses these issues to facilitate the contextualization and interpretation of human missense variation with unparalleled flexibility and ease of accessibility for use by the broadest range of researchers. By precalculating all possible variants in the human proteome it offers near instantaneous mapping between all relevant data types. It also combines data and analyses from a plethora of resources to bring together genomic, protein sequence and function annotations as well as structural insights and predictions to better understand the likely effect of missense variation in humans. It is offered as an intuitive web server https://www.ebi.ac.uk/protvar where data can be explored and downloaded, and can be accessed programmatically via an API.
Collapse
Affiliation(s)
| | - Prabhat Totoo
- EMBL-EBI, Wellcome Genome Campus, Hinxton CB10 1SD, Cambridgeshire, UK
| | | | - Jürgen Jänes
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Pedro Beltrao
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Maria J Martin
- EMBL-EBI, Wellcome Genome Campus, Hinxton CB10 1SD, Cambridgeshire, UK
| |
Collapse
|
9
|
Armirola-Ricaurte C, Morant L, Adant I, Hamed SA, Pipis M, Efthymiou S, Amor-Barris S, Atkinson D, Van de Vondel L, Tomic A, de Vriendt E, Zuchner S, Ghesquiere B, Hanna M, Houlden H, Lunn MP, Reilly MM, Rasic VM, Jordanova A. Biallelic variants in COX18 cause a mitochondrial disorder primarily manifesting as peripheral neuropathy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.03.24309787. [PMID: 39006432 PMCID: PMC11245062 DOI: 10.1101/2024.07.03.24309787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Defects in mitochondrial dynamics are a common cause of Charcot-Marie-Tooth disease (CMT), while primary deficiencies in the mitochondrial respiratory chain (MRC) are rare and atypical for this etiology. This study aims to report COX18 as a novel CMT-causing gene. This gene encodes an assembly factor of mitochondrial Complex IV (CIV) that translocates the C-terminal tail of MTCO2 across the mitochondrial inner membrane. Exome sequencing was performed in four affected individuals. The patients and available family members underwent thorough neurological and electrophysiological assessment. The impact of one of the identified variants on splicing, protein levels, and mitochondrial bioenergetics was investigated in patient-derived lymphoblasts. The functionality of the mutant protein was assessed using a Proteinase K protection assay and immunoblotting. Neuronal relevance of COX18 was assessed in a Drosophila melanogaster knockdown model. Exome sequencing coupled with homozygosity mapping revealed a homozygous splice variant c.435-6A>G in COX18 in two siblings with early-onset progressive axonal sensory-motor peripheral neuropathy. By querying external databases, we identified two additional families with rare deleterious biallelic variants in COX18 . All affected individuals presented with axonal CMT and some patients also exhibited central nervous system symptoms, such as dystonia and spasticity. Functional characterization of the c.435-6A>G variant demonstrated that it leads to the expression of an alternative transcript that lacks exon 2, resulting in a stable but defective COX18 isoform. The mutant protein impairs CIV assembly and activity, leading to a reduction in mitochondrial membrane potential. Downregulation of the COX18 homolog in Drosophila melanogaster displayed signs of neurodegeneration, including locomotor deficit and progressive axonal degeneration of sensory neurons. Our study presents genetic and functional evidence that supports COX18 as a newly identified gene candidate for autosomal recessive axonal CMT with or without central nervous system involvement. These findings emphasize the significance of peripheral neuropathy within the spectrum of primary mitochondrial disorders and the role of mitochondrial CIV in the development of CMT. Our research has important implications for the diagnostic workup of CMT patients.
Collapse
|
10
|
Hoffman J, Tan H, Sandoval-Cooper C, de Villiers K, Reed SM. GTExome: Modeling commonly expressed missense mutations in the human genome. PLoS One 2024; 19:e0303604. [PMID: 38814966 PMCID: PMC11139294 DOI: 10.1371/journal.pone.0303604] [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: 01/25/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024] Open
Abstract
A web application, GTExome, is described that quickly identifies, classifies, and models missense mutations in commonly expressed human proteins. GTExome can be used to categorize genomic mutation data with tissue specific expression data from the Genotype-Tissue Expression (GTEx) project. Commonly expressed missense mutations in proteins from a wide range of tissue types can be selected and assessed for modeling suitability. Information about the consequences of each mutation is provided to the user including if disulfide bonds, hydrogen bonds, or salt bridges are broken, buried prolines introduced, buried charges are created or lost, charge is swapped, a buried glycine is replaced, or if the residue that would be removed is a proline in the cis configuration. Also, if the mutation site is in a binding pocket the number of pockets and their volumes are reported. The user can assess this information and then select from available experimental or computationally predicted structures of native proteins to create, visualize, and download a model of the mutated protein using Fast and Accurate Side-chain Protein Repacking (FASPR). For AlphaFold modeled proteins, confidence scores for native proteins are provided. Using this tool, we explored a set of 9,666 common missense mutations from a variety of tissues from GTEx and show that most mutations can be modeled using this tool to facilitate studies of protein-protein and protein-drug interactions. The open-source tool is freely available at https://pharmacogenomics.clas.ucdenver.edu/gtexome/.
Collapse
Affiliation(s)
- Jill Hoffman
- Computational Bioscience, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Henry Tan
- Department of Chemistry, University of Colorado Denver, Denver, CO, United States of America
| | - Clara Sandoval-Cooper
- Department of Chemistry, University of Colorado Denver, Denver, CO, United States of America
| | - Kaelyn de Villiers
- Department of Chemistry, University of Colorado Denver, Denver, CO, United States of America
| | - Scott M. Reed
- Department of Chemistry, University of Colorado Denver, Denver, CO, United States of America
| |
Collapse
|
11
|
Colbert BM, Lanting C, Smeal M, Blanton S, Dykxhoorn DM, Tang PC, Getchell RL, Velde H, Fehrmann M, Thorpe R, Chapagain P, Elkhaligy H, Kremer H, Yntema H, Haer-Wigman L, Redfield S, Sun T, Bruijn S, Plomp A, Goderie T, van de Kamp J, Free RH, Wassink-Ruiter JK, Widdershoven J, Vanhoutte E, Rotteveel L, Kriek M, van Dooren M, Hoefsloot L, de Gier HHW, Schaefer A, Kolbe D, Azaiez H, Rabie G, Aburayyan A, Kawas M, Kanaan M, Holder J, Usami SI, Chen Z, Dai P, Holt J, Nelson R, Choi BY, Shearer E, Smith RJH, Pennings R, Liu XZ. The natural history and genotype-phenotype correlations of TMPRSS3 hearing loss: an international, multi-center, cohort analysis. Hum Genet 2024; 143:721-734. [PMID: 38691166 PMCID: PMC11098735 DOI: 10.1007/s00439-024-02648-3] [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: 10/18/2023] [Accepted: 01/21/2024] [Indexed: 05/03/2024]
Abstract
TMPRSS3-related hearing loss presents challenges in correlating genotypic variants with clinical phenotypes due to the small sample sizes of previous studies. We conducted a cross-sectional genomics study coupled with retrospective clinical phenotype analysis on 127 individuals. These individuals were from 16 academic medical centers across 6 countries. Key findings revealed 47 unique TMPRSS3 variants with significant differences in hearing thresholds between those with missense variants versus those with loss-of-function genotypes. The hearing loss progression rate for the DFNB8 subtype was 0.3 dB/year. Post-cochlear implantation, an average word recognition score of 76% was observed. Of the 51 individuals with two missense variants, 10 had DFNB10 with profound hearing loss. These 10 all had at least one of 4 TMPRSS3 variants predicted by computational modeling to be damaging to TMPRSS3 structure and function. To our knowledge, this is the largest study of TMPRSS3 genotype-phenotype correlations. We find significant differences in hearing thresholds, hearing loss progression, and age of presentation, by TMPRSS3 genotype and protein domain affected. Most individuals with TMPRSS3 variants perform well on speech recognition tests after cochlear implant, however increased age at implant is associated with worse outcomes. These findings provide insight for genetic counseling and the on-going design of novel therapeutic approaches.
Collapse
Affiliation(s)
- Brett M Colbert
- Department of Otolaryngology, University of Miami Miller School of Medicine, 1120 NW 14th Street, 5th Floor, Miami, FL, 33136, USA
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, USA
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, USA
| | - Cris Lanting
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Molly Smeal
- Department of Otolaryngology, University of Miami Miller School of Medicine, 1120 NW 14th Street, 5th Floor, Miami, FL, 33136, USA
| | - Susan Blanton
- Department of Otolaryngology, University of Miami Miller School of Medicine, 1120 NW 14th Street, 5th Floor, Miami, FL, 33136, USA
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, USA
| | - Derek M Dykxhoorn
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, USA
| | - Pei-Ciao Tang
- Department of Otolaryngology, University of Miami Miller School of Medicine, 1120 NW 14th Street, 5th Floor, Miami, FL, 33136, USA
| | - Richard L Getchell
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, USA
| | - Hedwig Velde
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mirthe Fehrmann
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ryan Thorpe
- Department of Otolaryngology, University of Iowa, Iowa City, USA
| | - Prem Chapagain
- Department of Physics and Biomolecular Sciences Institute, Florida International University, Miami, USA
| | - Heidy Elkhaligy
- Department of Physics and Biomolecular Sciences Institute, Florida International University, Miami, USA
| | - Hannie Kremer
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Helger Yntema
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lonneke Haer-Wigman
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Tieqi Sun
- Boston Children's Hospital, Boston, USA
| | - Saskia Bruijn
- Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Astrid Plomp
- Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Thadé Goderie
- Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Rolien H Free
- Groningen University Medical Center, Groningen, The Netherlands
| | | | | | - Els Vanhoutte
- Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | | | | | | | - Amanda Schaefer
- Department of Otolaryngology, University of Iowa, Iowa City, USA
| | - Diana Kolbe
- Department of Otolaryngology, University of Iowa, Iowa City, USA
| | - Hela Azaiez
- Department of Otolaryngology, University of Iowa, Iowa City, USA
| | - Grace Rabie
- Hereditary Research Laboratory and Department of Life Sciences, Bethlehem University, Bethlehem, Palestine
| | | | - Mariana Kawas
- Hereditary Research Laboratory and Department of Life Sciences, Bethlehem University, Bethlehem, Palestine
| | - Moien Kanaan
- Hereditary Research Laboratory and Department of Life Sciences, Bethlehem University, Bethlehem, Palestine
| | | | | | - Zhengyi Chen
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary and Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, USA
| | - Pu Dai
- PLA General Hospital, Beijing, China
| | | | - Rick Nelson
- Department of Otolaryngology, Indiana University School of Medicine, Indianapolis, USA
| | - Byung Yoon Choi
- Seoul National University Bundang Hospital, Seongnam, South Korea
| | | | | | - Ronald Pennings
- Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, 1120 NW 14th Street, 5th Floor, Miami, FL, 33136, USA.
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, USA.
| |
Collapse
|
12
|
Lyu Y, Wang T, Lin M, Qi F. A rare inherited homozygous missense variant in PLA2G6 influences susceptibility to infantile neuroaxonal dystrophy: a case report. Transl Pediatr 2024; 13:484-491. [PMID: 38590380 PMCID: PMC10998992 DOI: 10.21037/tp-23-568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/01/2024] [Indexed: 04/10/2024] Open
Abstract
Background Infantile neuroaxonal dystrophy (INAD) is an ultra-rare early-onset autosomal recessive neurodegenerative disorder due to PLA2G6 variants. The clinical symptoms of INAD patients display considerable diversity, and many PLA2G6 variants are still not thoroughly investigated in relation to their associated clinical presentations. Case Description A 16-month-old boy was admitted to our hospital due to regression of acquired motor and speech abilities that had persisted for 4 months. The patient was born to a healthy consanguineous couple after 41 weeks of pregnancy and natural delivery. Before 12 months old, he had normal motor development milestones. On admission, he also showed astasia-abasia, weakness of distal muscles, and diminished patellar tendon reflex. Brain magnetic resonance imaging (MRI) revealed cerebellar atrophy. Auditory brainstem response (ABR) indicated moderately severe hearing loss. With chromosome microarray analysis (CMA), we identified several copy number-neutral regions of runs of homozygosity (ROH) in the patient. Whole-exome sequencing (WES) further revealed that the patient harbored a homozygous missense variant NM_003560.2: c.1778C>T, p.Pro593Leu (rs1451486649) in the PLA2G6 gene. In the patient's asymptomatic parents and brother, the PLA2G6 c.1778C>T variant stayed in heterozygous status as confirmed by Sanger sequencing. The patient was finally diagnosed with INAD. Conclusions We report an INAD child with a rare PLA2G6 c.1778C>T homozygous missense variant and associated clinical symptoms. The family-based cosegregation analysis reveals that the PLA2G6 c.1778C>T homozygous variant contributes to the pathogenesis of INAD.
Collapse
Affiliation(s)
- Yongxue Lyu
- Department of Pediatric Health Care, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| | - Tao Wang
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Dian Diagnostics Group Co., Ltd., Hangzhou, China
| | - Meifang Lin
- Department of Pediatric Health Care, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| | - Fengfeng Qi
- Department of Pediatric Health Care, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| |
Collapse
|
13
|
Al-Kasbi G, Al-Murshedi F, Al-Futaisi A, Al-Jabry T, Zadjali F, Al-Yahyaee S, Al-Maawali A. Revisiting Exome Data Identified Missed Splice Site Variant of the Asparagine Synthetase ( ASNS ) Gene. J Pediatr Genet 2024; 13:1-5. [PMID: 38567172 PMCID: PMC10984708 DOI: 10.1055/s-0042-1757193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/21/2022] [Indexed: 04/04/2024]
Abstract
Next-generation sequencing, such as whole-exome sequencing (WES), is increasingly used in the study of Mendelian disorders, yet many are reported as "negative." Inappropriate variant annotation and filtering steps are reasons for missing the molecular diagnosis. Noncoding variants, including splicing mutations, are examples of variants that can be overlooked. Herein, we report a family of four affected newborns, and all presented with severe congenital microcephaly. Initial research WES analysis identified a damaging homozygous variant in NME1 gene as a possible cause of primary microcephaly phenotype in these patients. However, reanalysis of the exome data uncovered a biallelic splice site variant in asparagine synthetase gene which seems to be the possible cause of the phenotype in these patients. This study highlights the importance of revisiting the exome data and the issue of "negative" exome and the afterward approaches to identify and prove new candidate genes.
Collapse
Affiliation(s)
- Ghalia Al-Kasbi
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Fathiya Al-Murshedi
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
- Genetic and Developmental Medicine Clinic, Sultan Qaboos University Hospital, Muscat, Oman
| | - Amna Al-Futaisi
- Department of Child Health, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Tariq Al-Jabry
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Fahad Zadjali
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Said Al-Yahyaee
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Almundher Al-Maawali
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
- Genetic and Developmental Medicine Clinic, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
14
|
Iwata-Otsubo A, Skraban CM, Yoshimura A, Sakata T, Alves CAP, Fiordaliso SK, Kuroda Y, Vengoechea J, Grochowsky A, Ernste P, Lulis L, Nesbitt A, Tayoun AA, Gray C, Towne MC, Radtke K, Normand EA, Rhodes L, Seiler C, Shirahige K, Izumi K. Biallelic variants in GTF3C5, a regulator of RNA polymerase III-mediated transcription, cause a multisystem developmental disorder. Hum Genet 2024; 143:437-453. [PMID: 38520561 DOI: 10.1007/s00439-024-02656-3] [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: 11/22/2023] [Accepted: 02/13/2024] [Indexed: 03/25/2024]
Abstract
General transcription factor IIIC subunit 5 (GTF3C5) encodes transcription factor IIIC63 (TFIIIC63). It binds to DNA to recruit another transcription factor, TFIIIB, and RNA polymerase III (Pol III) to mediate the transcription of small noncoding RNAs, such as tRNAs. Here, we report four individuals from three families presenting with a multisystem developmental disorder phenotype with biallelic variants in GTF3C5. The overlapping features include growth retardation, developmental delay, intellectual disability, dental anomalies, cerebellar malformations, delayed bone age, skeletal anomalies, and facial dysmorphism. Using lymphoblastoid cell lines (LCLs) from two affected individuals, we observed a reduction in TFIIIC63 protein levels compared to control LCLs. Genome binding of TFIIIC63 protein is also reduced in LCL from one of the affected individuals. Additionally, approximately 40% of Pol III binding regions exhibited reduction in the level of Pol III occupancy in the mutant genome relative to the control, while approximately 54% of target regions showed comparable levels of Pol III occupancy between the two, indicating partial impairment of Pol III occupancy in the mutant genome. Yeasts with subject-specific variants showed temperature sensitivity and impaired growth, supporting the notion that the identified variants have deleterious effects. gtf3c5 mutant zebrafish showed developmental defects, including a smaller body, head, and eyes. Taken together, our data show that GTF3C5 plays an important role in embryonic development, and that biallelic variants in this gene cause a multisystem developmental disorder. Our study adds GTF3C5-related disorder to the growing list of genetic disorders associated with Pol III transcription machinery.
Collapse
Affiliation(s)
- Aiko Iwata-Otsubo
- Division of Human Genetics/Roberts Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Pathology, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA.
| | - Cara M Skraban
- Division of Human Genetics/Roberts Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Atsunori Yoshimura
- Laboratory of Genome Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Toyonori Sakata
- Laboratory of Genome Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Cesar Augusto P Alves
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Sarah K Fiordaliso
- Division of Human Genetics/Roberts Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Yukiko Kuroda
- Division of Human Genetics/Roberts Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jaime Vengoechea
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Angela Grochowsky
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Paige Ernste
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Invitae, San Francisco, CA, 94103, USA
| | - Lauren Lulis
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Addie Nesbitt
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Veritas Genetics, Danvers, MA, 01923, USA
| | - Ahmad Abou Tayoun
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Genomics Center of Excellence, Al Jalila Children's Specialty Hospital, Dubai Health, Center for Genomic Discovery, Mohammed Bin Rashid University, Dubai Health, UAE
| | - Christopher Gray
- Division of Human Genetics/Roberts Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | | | | | | | - Christoph Seiler
- Zebrafish Core, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Katsuhiko Shirahige
- Laboratory of Genome Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Kosuke Izumi
- Division of Human Genetics/Roberts Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Pediatrics, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Laboratory of Rare Disease Research, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan.
- Division of Genetics and Metabolism, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-8573, USA.
| |
Collapse
|
15
|
Kulshreshtha A, Bhatnagar S. Structural effect of the H992D/H418D mutation of angiotensin-converting enzyme in the Indian population: implications for health and disease. J Biomol Struct Dyn 2024:1-18. [PMID: 38411559 DOI: 10.1080/07391102.2024.2321246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
The Non synonymous SNPs (nsSNPs) of the renin-angiotensin-system (RAS) pathway, unique to the Indian population were investigated in view of its importance as an endocrine system. nsSNPs of the RAS pathway genes were mined from the IndiGenome database. Damaging nsSNPs were predicted using SIFT, PredictSNP, SNP and GO, Snap2 and Protein Variation Effect Analyzer. Loss of function was predicted based on protein stability change using I mutant, PremPS and CONSURF. The structural impact of the nsSNPs was predicted using HOPE and Missense3d followed by modeling, refinement, and energy minimization. Molecular Dynamics studies were carried out using Gromacsv2021.1. 23 Indian nsSNPs of the RAS pathway genes were selected for structural analysis and 8 were predicted to be damaging. Further sequence analysis showed that HEMGH zinc binding motif changes to HEMGD in somatic ACE-C domain (sACE-C) H992D and Testis ACE (tACE) H418D resulted in loss of zinc coordination, which is essential for enzymatic activity in this metalloprotease. There was a loss of internal interactions around the zinc coordination residues in the protein structural network. This was also confirmed by Principal Component Analysis, Free Energy Landscape and residue contact maps. Both mutations lead to broadening of the AngI binding cavity. The H992D mutation in sACE-C is likely to be favorable for cardiovascular health, but may lead to renal abnormalities with secondary impact on the heart. H418D in tACE is potentially associated with male infertility.
Collapse
Affiliation(s)
- Akanksha Kulshreshtha
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, India
| | - Sonika Bhatnagar
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, India
| |
Collapse
|
16
|
Hanna G, Khanna T, Islam SA, David A, Sternberg MJE. Missense3D-TM: Predicting the Effect of Missense Variants in Helical Transmembrane Protein Regions Using 3D Protein Structures. J Mol Biol 2024; 436:168374. [PMID: 38182301 PMCID: PMC7617522 DOI: 10.1016/j.jmb.2023.168374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/29/2023] [Accepted: 11/20/2023] [Indexed: 01/07/2024]
Abstract
Variant effect predictors assess if a substitution is pathogenic or benign. Most predictors, including those that are structure-based, are designed for globular proteins in aqueous environments and do not consider that the variant residue is located within the membrane. We report Missense3D-TM that provides a structure-based assessment of the impact of a missense variant located within a membrane. On a dataset of 2,078 pathogenic and 1,060 benign variants, spanning 711 proteins from 706 structures, Missense3D-TM achieved an accuracy of 66%, Mathews correlation coefficient of 0.37, sensitivity of 58% and specificity of 81%. Missense3D-TM performed similarly to mCSM-membrane: accuracy 66% vs 61% (p = 0.02) on an unbalanced test set and 70% vs 67% (p = 0.20) on a balanced test set. The Missense3D-TM website provides an analysis of the structural effects of the variant along with its predicted position within the membrane. The web server is available at http://missense3d.bc.ic.ac.uk/.
Collapse
Affiliation(s)
- Gordon Hanna
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Tarun Khanna
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Suhail A Islam
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Alessia David
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK.
| | - Michael J E Sternberg
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
17
|
Bamezai S, Maresca di Serracapriola G, Morris F, Hildebrandt R, Amil MAS, Ledesma‐Amaro R. Protein engineering in the computational age: An open source framework for exploring mutational landscapes in silico. ENGINEERING BIOLOGY 2023; 7:29-38. [PMID: 38094241 PMCID: PMC10715127 DOI: 10.1049/enb2.12028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/04/2023] [Accepted: 10/25/2023] [Indexed: 10/16/2024] Open
Abstract
The field of protein engineering has seen tremendous expansion in the last decade, with researchers developing novel proteins with specialised functionalities for a range of uses, from drug discovery to industrial biotechnology. The emergence of computational tools and high-throughput screening technology has substantially sped up the process of protein engineering. However, much of the expertise required to engage in such projects is still concentrated in the hands of a few specialised individuals, including computational biologists and structural biochemists. The international Genetically Engineered Machine (iGEM) competition represents a platform for undergraduate students to innovate in synthetic biology. Yet, due to their complexity, arduous protein engineering projects are hindered by the resources available and strict timelines of the competition. The authors highlight how the 2022 iGEM Team, 'Sporadicate', set out to develop InFinity 1.0, a computational framework for increased accessibility to effective protein engineering, hoping to increase awareness and accessibility to novel in silico tools.
Collapse
Affiliation(s)
- Shirin Bamezai
- Department of Bioengineering and Imperial College Centre for Synthetic BiologyImperial College LondonLondonUK
| | | | - Freya Morris
- Department of Bioengineering and Imperial College Centre for Synthetic BiologyImperial College LondonLondonUK
| | | | | | - Rodrigo Ledesma‐Amaro
- Department of Bioengineering and Imperial College Centre for Synthetic BiologyImperial College LondonLondonUK
| |
Collapse
|
18
|
Hoffman J, Tan H, Sandoval-Cooper C, de Villiers K, Reed SM. GTExome: Modeling commonly expressed missense mutations in the human genome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567143. [PMID: 38014287 PMCID: PMC10680684 DOI: 10.1101/2023.11.14.567143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
A web application, GTExome, is described that quickly identifies, classifies, and models missense mutations in commonly expressed human proteins. GTExome can be used to categorize genomic mutation data with tissue specific expression data from the Genotype-Tissue Expression (GTEx) project. Commonly expressed missense mutations in proteins from a wide range of tissue types can be selected and assessed for modeling suitability. Information about the consequences of each mutation is provided to the user including if disulfide bonds, hydrogen bonds, or salt bridges are broken, buried prolines introduced, buried charges are created or lost, charge is swapped, a buried glycine is replaced, or if the residue that would be removed is a proline in the cis configuration. Also, if the mutation site is in a binding pocket the number of pockets and their volumes are reported. The user can assess this information and then select from available experimental or computationally predicted structures of native proteins to create, visualize, and download a model of the mutated protein using Fast and Accurate Side-chain Protein Repacking (FASPR). For AlphaFold modeled proteins, confidence scores for native proteins are provided. Using this tool, we explored a set of 9,666 common missense mutations from a variety of tissues from GTEx and show that most mutations can be modeled using this tool to facilitate studies of protein-protein and protein-drug interactions. The open-source tool is freely available at https://pharmacogenomics.clas.ucdenver.edu/gtexome/.
Collapse
Affiliation(s)
| | | | | | | | - Scott M. Reed
- Department of Chemistry, Department of Chemistry, University of Colorado Denver, 1151 Arapahoe St., Denver, CO 80204 USA
| |
Collapse
|
19
|
Malik A, Sharma D, Aguirre-Gamboa R, McGrath S, Zabala S, Weber C, Jabri B. Epithelial IFNγ signalling and compartmentalized antigen presentation orchestrate gut immunity. Nature 2023; 623:1044-1052. [PMID: 37993709 PMCID: PMC11361632 DOI: 10.1038/s41586-023-06721-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/06/2023] [Indexed: 11/24/2023]
Abstract
All nucleated cells express major histocompatibility complex I and interferon-γ (IFNγ) receptor1, but an epithelial cell-specific function of IFNγ signalling or antigen presentation by means of major histocompatibility complex I has not been explored. We show here that on sensing IFNγ, colonic epithelial cells productively present pathogen and self-derived antigens to cognate intra-epithelial T cells, which are critically located at the epithelial barrier. Antigen presentation by the epithelial cells confers extracellular ATPase expression in cognate intra-epithelial T cells, which limits the accumulation of extracellular adenosine triphosphate and consequent activation of the NLRP3 inflammasome in tissue macrophages. By contrast, antigen presentation by the tissue macrophages alongside inflammasome-associated interleukin-1α and interleukin-1β production promotes a pathogenic transformation of CD4+ T cells into granulocyte-macrophage colony-stimulating-factor (GM-CSF)-producing T cells in vivo, which promotes colitis and colorectal cancer. Taken together, our study unravels critical checkpoints requiring IFNγ sensing and antigen presentation by epithelial cells that control the development of pathogenic CD4+ T cell responses in vivo.
Collapse
Affiliation(s)
- Ankit Malik
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA.
| | - Deepika Sharma
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Raúl Aguirre-Gamboa
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Shaina McGrath
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Sarah Zabala
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Christopher Weber
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
- Department of Pathology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Bana Jabri
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
20
|
Pelletier D, Chong AL, Wu M, Witkowski L, Albert S, Sabbaghian N, Fabian M, Foulkes W. DICER1 platform domain missense variants inhibit miRNA biogenesis and lead to tumor susceptibility. NAR Cancer 2023; 5:zcad030. [PMID: 37333613 PMCID: PMC10273190 DOI: 10.1093/narcan/zcad030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/10/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Abstract
The endoribonuclease DICER1 plays an essential role in the microRNA (miRNA) biogenesis pathway, cleaving precursor miRNA (pre-miRNA) stem-loops to generate mature single-stranded miRNAs. Germline pathogenic variants (GPVs) in DICER1 result in DICER1 tumor predisposition syndrome (DTPS), a mainly childhood-onset tumor susceptibility disorder. Most DTPS-causing GPVs are nonsense or frameshifting, with tumor development requiring a second somatic missense hit that impairs the DICER1 RNase IIIb domain. Interestingly, germline DICER1 missense variants that cluster in the DICER1 Platform domain have been identified in some persons affected by tumors that also associate with DTPS. Here, we demonstrate that four of these Platform domain variants prevent DICER1 from producing mature miRNAs and as a result impair miRNA-mediated gene silencing. Importantly, we show that in contrast to canonical somatic missense variants that alter DICER1 cleavage activity, DICER1 proteins harboring these Platform variants fail to bind to pre-miRNA stem-loops. Taken together, this work sheds light upon a unique subset of GPVs causing DTPS and provides new insights into how alterations in the DICER1 Platform domain can impact miRNA biogenesis.
Collapse
Affiliation(s)
- Dylan Pelletier
- Department of Human Genetics, Medicine, McGill University, Montreal, QC, Canada
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Anne-Laure Chong
- Department of Human Genetics, Medicine, McGill University, Montreal, QC, Canada
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
| | - Mona Wu
- Department of Human Genetics, Medicine, McGill University, Montreal, QC, Canada
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
| | - Leora Witkowski
- Department of Human Genetics, Medicine, McGill University, Montreal, QC, Canada
| | - Sophie Albert
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
| | - Nelly Sabbaghian
- Department of Human Genetics, Medicine, McGill University, Montreal, QC, Canada
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
| | - Marc R Fabian
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - William D Foulkes
- Department of Human Genetics, Medicine, McGill University, Montreal, QC, Canada
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
21
|
Valencia AM, Sankar A, van der Sluijs PJ, Satterstrom FK, Fu J, Talkowski ME, Vergano SAS, Santen GWE, Kadoch C. Landscape of mSWI/SNF chromatin remodeling complex perturbations in neurodevelopmental disorders. Nat Genet 2023; 55:1400-1412. [PMID: 37500730 PMCID: PMC10412456 DOI: 10.1038/s41588-023-01451-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 06/20/2023] [Indexed: 07/29/2023]
Abstract
DNA sequencing-based studies of neurodevelopmental disorders (NDDs) have identified a wide range of genetic determinants. However, a comprehensive analysis of these data, in aggregate, has not to date been performed. Here, we find that genes encoding the mammalian SWI/SNF (mSWI/SNF or BAF) family of ATP-dependent chromatin remodeling protein complexes harbor the greatest number of de novo missense and protein-truncating variants among nuclear protein complexes. Non-truncating NDD-associated protein variants predominantly disrupt the cBAF subcomplex and cluster in four key structural regions associated with high disease severity, including mSWI/SNF-nucleosome interfaces, the ATPase-core ARID-armadillo repeat (ARM) module insertion site, the Arp module and DNA-binding domains. Although over 70% of the residues perturbed in NDDs overlap with those mutated in cancer, ~60% of amino acid changes are NDD-specific. These findings provide a foundation to functionally group variants and link complex aberrancies to phenotypic severity, serving as a resource for the chromatin, clinical genetics and neurodevelopment communities.
Collapse
Affiliation(s)
- Alfredo M Valencia
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Chemical Biology Program, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Akshay Sankar
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - F Kyle Satterstrom
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Jack Fu
- Massachusetts General Hospital, Boston, MA, USA
| | - Michael E Talkowski
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Samantha A Schrier Vergano
- Children's Hospital of the King's Daughters, Norfolk, Virginia, USA
- Department of Pediatrics, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Gijs W E Santen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Cigall Kadoch
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
22
|
Pennica C, Hanna G, Islam SA, Sternberg MJE, David A. Missense3D-PPI: A Web Resource to Predict the Impact of Missense Variants at Protein Interfaces Using 3D Structural Data. J Mol Biol 2023; 435:168060. [PMID: 37356905 PMCID: PMC7617523 DOI: 10.1016/j.jmb.2023.168060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
In 2019, we released Missense3D which identifies stereochemical features that are disrupted by a missense variant, such as introducing a buried charge. Missense3D analyses the effect of a missense variant on a single structure and thus may fail to identify as damaging surface variants disrupting a protein interface i.e., a protein-protein interaction (PPI) site. Here we present Missense3D-PPI designed to predict missense variants at PPI interfaces. Our development dataset comprised of 1,279 missense variants (pathogenic n = 733, benign n = 546) in 434 proteins and 545 experimental structures of PPI complexes. Benchmarking of Missense3D-PPI was performed after dividing the dataset in training (320 benign and 320 pathogenic variants) and testing (226 benign and 413 pathogenic). Structural features affecting PPI, such as disruption of interchain bonds and introduction of unbalanced charged interface residues, were analysed to assess the impact of the variant at PPI. The performance of Missense3D-PPI was superior to that of Missense3D: sensitivity 44 % versus 8% and accuracy 58% versus 40%, p = 4.23 × 10-16. However, the specificity of Missense3D-PPI was lower compared to Missense3D (84% versus 98%). On our dataset, Missense3D-PPI's accuracy was superior to BeAtMuSiC (p = 3.4 × 10-5), mCSM-PPI2 (p = 1.5 × 10-12) and MutaBind2 (p = 0.0025). Missense3D-PPI represents a valuable tool for predicting the structural effect of missense variants on biological protein networks and is available at the Missense3D web portal (http://missense3d.bc.ic.ac.uk).
Collapse
Affiliation(s)
- Cecilia Pennica
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Gordon Hanna
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Suhail A Islam
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Michael J E Sternberg
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Alessia David
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
23
|
Leggatt G, Cheng G, Narain S, Briseño-Roa L, Annereau JP, Gast C, Gilbert RD, Ennis S. A genotype-to-phenotype approach suggests under-reporting of single nucleotide variants in nephrocystin-1 (NPHP1) related disease (UK 100,000 Genomes Project). Sci Rep 2023; 13:9369. [PMID: 37296294 PMCID: PMC10256716 DOI: 10.1038/s41598-023-32169-4] [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: 08/26/2022] [Accepted: 03/23/2023] [Indexed: 06/12/2023] Open
Abstract
Autosomal recessive whole gene deletions of nephrocystin-1 (NPHP1) result in abnormal structure and function of the primary cilia. These deletions can result in a tubulointerstitial kidney disease known as nephronophthisis and retinal (Senior-Løken syndrome) and neurological (Joubert syndrome) diseases. Nephronophthisis is a common cause of end-stage kidney disease (ESKD) in children and up to 1% of adult onset ESKD. Single nucleotide variants (SNVs) and small insertions and deletions (Indels) have been less well characterised. We used a gene pathogenicity scoring system (GenePy) and a genotype-to-phenotype approach on individuals recruited to the UK Genomics England (GEL) 100,000 Genomes Project (100kGP) (n = 78,050). This approach identified all participants with NPHP1-related diseases reported by NHS Genomics Medical Centres and an additional eight participants. Extreme NPHP1 gene scores, often underpinned by clear recessive inheritance, were observed in patients from diverse recruitment categories, including cancer, suggesting the possibility of a more widespread disease than previously appreciated. In total, ten participants had homozygous CNV deletions with eight homozygous or compound heterozygous with SNVs. Our data also reveals strong in-silico evidence that approximately 44% of NPHP1 related disease may be due to SNVs with AlphaFold structural modelling evidence for a significant impact on protein structure. This study suggests historical under-reporting of SNVS in NPHP1 related diseases compared with CNVs.
Collapse
Affiliation(s)
- Gary Leggatt
- University of Southampton, Duthie Building (MP 808), Southampton General Hospital, Tremona Road Shirley, Southampton, SO16 6YD, UK.
- Wessex Kidney Centre, Portsmouth Hospitals University NHS Trust, Southwick Hill Road, Cosham, Portsmouth, PO6 3LY, UK.
- University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Tremona Road Shirley, Southampton, SO16 6YD, UK.
| | - Guo Cheng
- University of Southampton, Duthie Building (MP 808), Southampton General Hospital, Tremona Road Shirley, Southampton, SO16 6YD, UK
| | - Sumit Narain
- University of Southampton, Duthie Building (MP 808), Southampton General Hospital, Tremona Road Shirley, Southampton, SO16 6YD, UK
| | - Luis Briseño-Roa
- Medetia, Imagine Institute for Genetic Diseases, 24 Boulevard du Montparnasse, 75015, Paris, France
| | - Jean-Philippe Annereau
- Medetia, Imagine Institute for Genetic Diseases, 24 Boulevard du Montparnasse, 75015, Paris, France
| | - Christine Gast
- University of Southampton, Duthie Building (MP 808), Southampton General Hospital, Tremona Road Shirley, Southampton, SO16 6YD, UK
- Wessex Kidney Centre, Portsmouth Hospitals University NHS Trust, Southwick Hill Road, Cosham, Portsmouth, PO6 3LY, UK
| | - Rodney D Gilbert
- University of Southampton, Duthie Building (MP 808), Southampton General Hospital, Tremona Road Shirley, Southampton, SO16 6YD, UK
- Southampton Children's Hospital, Southampton General Hospital, Tremona Road Shirley, Southampton, SO16 6YD, UK
| | - Sarah Ennis
- University of Southampton, Duthie Building (MP 808), Southampton General Hospital, Tremona Road Shirley, Southampton, SO16 6YD, UK
| |
Collapse
|
24
|
David A, Sternberg MJE. Protein structure-based evaluation of missense variants: Resources, challenges and future directions. Curr Opin Struct Biol 2023; 80:102600. [PMID: 37126977 DOI: 10.1016/j.sbi.2023.102600] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 05/03/2023]
Abstract
We provide an overview of the methods that can be used for protein structure-based evaluation of missense variants. The algorithms can be broadly divided into those that calculate the difference in free energy (ΔΔG) between the wild type and variant structures and those that use structural features to predict the damaging effect of a variant without providing a ΔΔG. A wide range of machine learning approaches have been employed to develop those algorithms. We also discuss challenges and opportunities for variant interpretation in view of the recent breakthrough in three-dimensional structural modelling using deep learning.
Collapse
Affiliation(s)
- Alessia David
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.
| | - Michael J E Sternberg
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
25
|
Li C, Hou I, Ma M, Wang G, Bai Y, Liu X. Orthogonal analysis of variants in APOE gene using in-silico approaches reveals novel disrupting variants. FRONTIERS IN BIOINFORMATICS 2023; 3:1122559. [PMID: 37091907 PMCID: PMC10117898 DOI: 10.3389/fbinf.2023.1122559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction: Alzheimer's disease (AD) is one of the most prominent medical conditions in the world. Understanding the genetic component of the disease can greatly advance our knowledge regarding its progression, treatment and prognosis. Single amino-acid variants (SAVs) in the APOE gene have been widely investigated as a risk factor for AD Studies, including genome-wide association studies, meta-analysis based studies, and in-vivo animal studies, were carried out to investigate the functional importance and pathogenesis potential of APOE SAVs. However, given the high cost of such large-scale or experimental studies, there are only a handful of variants being reported that have definite explanations. The recent development of in-silico analytical approaches, especially large-scale deep learning models, has opened new opportunities for us to probe the structural and functional importance of APOE variants extensively. Method: In this study, we are taking an ensemble approach that simultaneously uses large-scale protein sequence-based models, including Evolutionary Scale Model and AlphaFold, together with a few in-silico functional prediction web services to investigate the known and possibly disease-causing SAVs in APOE and evaluate their likelihood of being functional and structurally disruptive. Results: As a result, using an ensemble approach with little to no prior field-specific knowledge, we reported 5 SAVs in APOE gene to be potentially disruptive, one of which (C112R) was classificed by previous studies as a key risk factor for AD. Discussion: Our study provided a novel framework to analyze and prioritize the functional and structural importance of SAVs for future experimental and functional validation.
Collapse
Affiliation(s)
- Chang Li
- USF Genomics and College of Public Health, University of South Florida, Tampa, FL, United States
| | - Ian Hou
- The John Cooper School, The Woodlands, TX, United States
| | - Mingjia Ma
- Novi High School, Novi, MI, United States
| | - Grace Wang
- Del Norte High School, San Diego, CA, United States
| | - Yongsheng Bai
- Next-Gen Intelligent Science Training, Ann Arbor, MI, United States
- Department of Biology, Eastern Michigan University, Ypsilanti, MI, United States
| | - Xiaoming Liu
- USF Genomics and College of Public Health, University of South Florida, Tampa, FL, United States
| |
Collapse
|
26
|
Musolf AM, Haarman AEG, Luben RN, Ong JS, Patasova K, Trapero RH, Marsh J, Jain I, Jain R, Wang PZ, Lewis DD, Tedja MS, Iglesias AI, Li H, Cowan CS, Consortium for Refractive Error and Myopia (CREAM), Biino G, Klein AP, Duggal P, Mackey DA, Hayward C, Haller T, Metspalu A, Wedenoja J, Pärssinen O, Cheng CY, Saw SM, Stambolian D, Hysi PG, Khawaja AP, Vitart V, Hammond CJ, van Duijn CM, Verhoeven VJM, Klaver CCW, Bailey-Wilson JE. Rare variant analyses across multiethnic cohorts identify novel genes for refractive error. Commun Biol 2023; 6:6. [PMID: 36596879 PMCID: PMC9810640 DOI: 10.1038/s42003-022-04323-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/30/2022] [Indexed: 01/05/2023] Open
Abstract
Refractive error, measured here as mean spherical equivalent (SER), is a complex eye condition caused by both genetic and environmental factors. Individuals with strong positive or negative values of SER require spectacles or other approaches for vision correction. Common genetic risk factors have been identified by genome-wide association studies (GWAS), but a great part of the refractive error heritability is still missing. Some of this heritability may be explained by rare variants (minor allele frequency [MAF] ≤ 0.01.). We performed multiple gene-based association tests of mean Spherical Equivalent with rare variants in exome array data from the Consortium for Refractive Error and Myopia (CREAM). The dataset consisted of over 27,000 total subjects from five cohorts of Indo-European and Eastern Asian ethnicity. We identified 129 unique genes associated with refractive error, many of which were replicated in multiple cohorts. Our best novel candidates included the retina expressed PDCD6IP, the circadian rhythm gene PER3, and P4HTM, which affects eye morphology. Future work will include functional studies and validation. Identification of genes contributing to refractive error and future understanding of their function may lead to better treatment and prevention of refractive errors, which themselves are important risk factors for various blinding conditions.
Collapse
Affiliation(s)
- Anthony M Musolf
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, USA
| | - Annechien E G Haarman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robert N Luben
- MRC Epidemiology, University of Cambridge School of Clinical Medicine, Cambridge, UK
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Jue-Sheng Ong
- Statistical Genetics Laboratory, Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Karina Patasova
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Rolando Hernandez Trapero
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Joseph Marsh
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Ishika Jain
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, USA
| | - Riya Jain
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, USA
| | - Paul Zhiping Wang
- Institute for Biomedical Sciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Deyana D Lewis
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, USA
| | - Milly S Tedja
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adriana I Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hengtong Li
- Data Science Unit, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Cameron S Cowan
- Institute for Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | | | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of Italy, Pavia, Italy
| | - Alison P Klein
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Priya Duggal
- The Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - David A Mackey
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, WA, Australia
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Toomas Haller
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Juho Wedenoja
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Olavi Pärssinen
- Department of Ophthalmology, Central Hospital of Central Finland, Jyväskylä, Finland
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Ching-Yu Cheng
- Centre for Quantitative Medicine, DUKE-National University of Singapore, Singapore, Singapore
- Ocular Epidemiology Research Group, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Seang-Mei Saw
- Saw Swee Hock School of Public Health, National University Health Systems, National University of Singapore, Singapore, Singapore
- Myopia Research Group, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Dwight Stambolian
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Pirro G Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Anthony P Khawaja
- MRC Epidemiology, University of Cambridge School of Clinical Medicine, Cambridge, UK
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Christopher J Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | | | - Virginie J M Verhoeven
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Institute for Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.
- Department of Ophthalmology, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Joan E Bailey-Wilson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
Collaborators
Paul Nigel Baird, Amutha Barathi Veluchamy, Kathryn P Burdon, Harry Campbell, Li Jia Chen, Ching-Yu Cheng, Emily Y Chew, Jamie E Craig, Phillippa M Cumberland, Margaret M Deangelis, Cécile Delcourt, Xiaohu Ding, David M Evans, Qiao Fan, Maurizio Fossarello, Paul J Foster, Puya Gharahkhani, Adriana I Iglesias, Jeremy A Guggenheim, Xiaobo Guo, Xikun Han, Mingguang He, Alex W Hewitt, Quan V Hoang, Sudha K Iyengar, Jost B Jonas, Mika Kähönen, Jaakko Kaprio, Barbara E Klein, Jonathan H Lass, Kris Lee, Terho Lehtimäki, Deyana Lewis, Qing Li, Shi-Ming Li, Leo-Pekka Lyytikäinen, Stuart MacGregor, David A Mackey, Nicholas G Martin, Akira Meguro, Candace Middlebrooks, Masahiro Miyake, Nobuhisa Mizuki, Anthony Musolf, Stefan Nickels, Konrad Oexle, Chi Pui Pang, Andrew D Paterson, Craig Pennell, Norbert Pfeiffer, Ozren Polasek, Jugnoo S Rahi, Olli Raitakari, Igor Rudan, Srujana Sahebjada, Claire L Simpson, E-Shyong Tai, Milly S Tedja, J Willem L Tideman, Akitaka Tsujikawa, Ningli Wang, Wen Bin Wei, Cathy Williams, Katie M Williams, James F Wilson, Robert Wojciechowski, Ya Xing Wang, Kenji Yamashiro, Jason C S Yam, Maurice K H Yap, Seyhan Yazar, Shea Ping Yip, Terri L Young, Xiangtian Zhou,
Collapse
|
27
|
Chatterjee A, Sarkar K, Bank S, Ghosh S, Kumar Pal D, Saraf S, Wakle D, Roy B, Chakraborty S, Bankura B, Chattopadhyay D, Das M. Homozygous GRHPR C.494G>A mutation is deleterious that causes early onset of nephrolithiasis in West Bengal, India. Front Mol Biosci 2022; 9:1049620. [PMID: 36619171 PMCID: PMC9815608 DOI: 10.3389/fmolb.2022.1049620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Pediatric nephrolithiasis (NL) or Kidney stone disease (KSD) is an untethered topic in Asian population. In Western countries, the annual incidence of paediatric NL is around 6-10%. Here, we present data from West Bengal, India, on lower age (LA, 0-20 years) NL and its prevalence for the first time. To discover the mutations associated with KSD, twenty-four (18 + 6) rare LA-NL patients were selected for Whole Exome Sequencing (WES) and Sanger sequencing, respectively. It was found that GRHPR c. 494G>A mutation (MZ826703) is predominant in our study cohort. This specific homozygous mutation is functionally studied for the first time directly from human peripheral mononuclear cell (PBMC) samples. Using expression study with biochemical activity and computational analysis we assumed that the mutation is pathogenic with loss of function. Moreover, three genes, AGXT, HOGA1 and GRHPR with Novel variants known to cause hyperoxaluria were found frequently in the study cohort. Our study analyses the genes and variations that cause LA-NL, as well as the molecular function of the GRHPR mutation, which may serve as a clinical marker in the population of West Bengal, Eastern India.
Collapse
Affiliation(s)
| | - Kunal Sarkar
- Department of Zoology, University of Calcutta, Kolkata, India
| | - Sarbashri Bank
- Department of Zoology, University of Calcutta, Kolkata, India
| | - Sudakshina Ghosh
- Department of Zoology, Vidyasagar College for Women, Kolkata, India
| | - Dilip Kumar Pal
- Department of Urology, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Siddharth Saraf
- Department of Urology, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Dhansagar Wakle
- Department of Urology, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Bidyut Roy
- Human Genetics Unit, Indian Statistical Institute, Kolkata, India
| | | | - Biswabandhu Bankura
- Department of Zoology, University of Calcutta, Kolkata, India,Medical College, Kolkata, India
| | - Debprasad Chattopadhyay
- ICMR Virus Unit, ID & BG Hospital, Kolkata, India,ICMR-National Institute of Traditional Medicine, Belgavi, India,School of Health Sciences, NSHM Knowledge Campus, Kolkata, India,*Correspondence: Madhusudan Das, ; Debprasad Chattopadhyay,
| | - Madhusudan Das
- Department of Zoology, University of Calcutta, Kolkata, India,*Correspondence: Madhusudan Das, ; Debprasad Chattopadhyay,
| |
Collapse
|
28
|
Wang S, Jain A, Novales NA, Nashner AN, Tran F, Clarke CF. Predicting and Understanding the Pathology of Single Nucleotide Variants in Human COQ Genes. Antioxidants (Basel) 2022; 11:antiox11122308. [PMID: 36552517 PMCID: PMC9774615 DOI: 10.3390/antiox11122308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022] Open
Abstract
Coenzyme Q (CoQ) is a vital lipid that functions as an electron carrier in the mitochondrial electron transport chain and as a membrane-soluble antioxidant. Deficiencies in CoQ lead to metabolic diseases with a wide range of clinical manifestations. There are currently few treatments that can slow or stop disease progression. Primary CoQ10 deficiency can arise from mutations in any of the COQ genes responsible for CoQ biosynthesis. While many mutations in these genes have been identified, the clinical significance of most of them remains unclear. Here we analyzed the structural and functional impact of 429 human missense single nucleotide variants (SNVs) that give rise to amino acid substitutions in the conserved and functional regions of human genes encoding a high molecular weight complex known as the CoQ synthome (or Complex Q), consisting of the COQ3-COQ7 and COQ9 gene products. Using structures of COQ polypeptides, close homologs, and AlphaFold models, we identified 115 SNVs that are potentially pathogenic. Further biochemical characterizations in model organisms such as Saccharomyces cerevisiae are required to validate the pathogenicity of the identified SNVs. Collectively, our results will provide a resource for clinicians during patient diagnosis and guide therapeutic efforts toward combating primary CoQ10 deficiency.
Collapse
|
29
|
Varadi M, Anyango S, Appasamy SD, Armstrong D, Bage M, Berrisford J, Choudhary P, Bertoni D, Deshpande M, Leines GD, Ellaway J, Evans G, Gaborova R, Gupta D, Gutmanas A, Harrus D, Kleywegt GJ, Bueno WM, Nadzirin N, Nair S, Pravda L, Afonso MQL, Sehnal D, Tanweer A, Tolchard J, Abrams C, Dunlop R, Velankar S. PDBe and PDBe-KB: Providing high-quality, up-to-date and integrated resources of macromolecular structures to support basic and applied research and education. Protein Sci 2022; 31:e4439. [PMID: 36173162 PMCID: PMC9517934 DOI: 10.1002/pro.4439] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/26/2022]
Abstract
The archiving and dissemination of protein and nucleic acid structures as well as their structural, functional and biophysical annotations is an essential task that enables the broader scientific community to conduct impactful research in multiple fields of the life sciences. The Protein Data Bank in Europe (PDBe; pdbe.org) team develops and maintains several databases and web services to address this fundamental need. From data archiving as a member of the Worldwide PDB consortium (wwPDB; wwpdb.org), to the PDBe Knowledge Base (PDBe-KB; pdbekb.org), we provide data, data-access mechanisms, and visualizations that facilitate basic and applied research and education across the life sciences. Here, we provide an overview of the structural data and annotations that we integrate and make freely available. We describe the web services and data visualization tools we offer, and provide information on how to effectively use or even further develop them. Finally, we discuss the direction of our data services, and how we aim to tackle new challenges that arise from the recent, unprecedented advances in the field of structure determination and protein structure modeling.
Collapse
Affiliation(s)
- Mihaly Varadi
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Stephen Anyango
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Sri Devan Appasamy
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - David Armstrong
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Marcus Bage
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - John Berrisford
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Preeti Choudhary
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Damian Bertoni
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Mandar Deshpande
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Grisell Diaz Leines
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Joseph Ellaway
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Genevieve Evans
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Romana Gaborova
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Deepti Gupta
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Aleksandras Gutmanas
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Deborah Harrus
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Gerard J Kleywegt
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | | | - Nurul Nadzirin
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Sreenath Nair
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Lukas Pravda
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | | | - David Sehnal
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
- CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Ahsan Tanweer
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - James Tolchard
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Charlotte Abrams
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Roisin Dunlop
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| | - Sameer Velankar
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton
| |
Collapse
|
30
|
Mintoff D, Pace NP, Borg I. Interpreting the spectrum of gamma-secretase complex missense variation in the context of hidradenitis suppurativa—An in-silico study. Front Genet 2022; 13:962449. [PMID: 36118898 PMCID: PMC9478468 DOI: 10.3389/fgene.2022.962449] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Hidradenitis suppurativa (HS) is a disease of the pilosebaceous unit characterized by recurrent nodules, abscesses and draining tunnels with a predilection to intertriginous skin. The pathophysiology of HS is complex. However, it is known that inflammation and hyperkeratinization at the hair follicle play crucial roles in disease manifestation. Genetic and environmental factors are considered the main drivers of these two pathophysiological processes. Despite a considerable proportion of patients having a positive family history of disease, only a minority of patients suffering from HS have been found to harbor monogenic variants which segregate to affected kindreds. Most of these variants are in the ɣ secretase complex (GSC) protein-coding genes. In this manuscript, we set out to characterize the burden of missense pathogenic variants in healthy reference population using large scale genomic dataset thereby providing a standard for comparing genomic variation in GSC protein-coding genes in the HS patient cohort.
Collapse
Affiliation(s)
- Dillon Mintoff
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Nikolai P. Pace
- Centre for Molecular Biology and Biobanking, University of Malta, Msida, Malta
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- *Correspondence: Nikolai P. Pace,
| | - Isabella Borg
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Centre for Molecular Biology and Biobanking, University of Malta, Msida, Malta
- Department of Pathology, Mater Dei Hospital, Msida, Malta
| |
Collapse
|
31
|
Khan AS, Parvez N, Ahsan T, Shoily SS, Sajib AA. A comprehensive in silico exploration of the impacts of missense variants on two different conformations of human pirin protein. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2022; 46:225. [PMID: 35967515 PMCID: PMC9362109 DOI: 10.1186/s42269-022-00917-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Pirin, a member of the cupin superfamily, is an iron-binding non-heme protein. It acts as a coregulator of several transcription factors, especially the members of NFκB transcription factor family. Based on the redox state of its iron cofactor, it can assume two different conformations and thereby act as a redox sensor inside the nucleus. Previous studies suggested that pirin may be associated with cancer, inflammatory diseases as well as COVID-19 severities. Hence, it is important to explore the pathogenicity of its missense variants. In this study, we used a number of in silico tools to investigate the effects of missense variants of pirin on its structure, stability, metal cofactor binding affinity and interactions with partner proteins. In addition, we used protein dynamics simulation to elucidate the effects of selected variants on its dynamics. Furthermore, we calculated the frequencies of haplotypes containing pirin missense variants across five major super-populations (African, Admixed American, East Asian, European and South Asian). RESULTS Among a total of 153 missense variants of pirin, 45 were uniformly predicted to be pathogenic. Of these, seven variants can be considered for further experimental studies. Variants R59P and L116P were predicted to significantly destabilize and damage pirin structure, substantially reduce its affinity to its binding partners and alter pirin residue fluctuation profile via changing the flexibility of several key residues. Additionally, variants R59Q, F78V, G98D, V151D and L220P were found to impact pirin structure and function in multiple ways. As no haplotype was identified to be harboring more than one missense variant, further interrogation of the individual effects of these seven missense variants is highly recommended. CONCLUSIONS Pirin is involved in the transcriptional regulation of several genes and can play an important role in inflammatory responses. The variants predicted to be pathogenic in this study may thus contribute to a better understanding of the underlying molecular mechanisms of various inflammatory diseases. Future studies should be focused on clarifying if any of these variants can be used as disease biomarkers. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1186/s42269-022-00917-7.
Collapse
Affiliation(s)
- Auroni Semonti Khan
- Department of Genetic Engineering and Biotechnology, Jagannath University, Dhaka, 1100 Bangladesh
| | - Nahid Parvez
- Department of Genetic Engineering and Biotechnology, Jagannath University, Dhaka, 1100 Bangladesh
| | - Tamim Ahsan
- Molecular Biotechnology Division, National Institute of Biotechnology, Savar, Dhaka, 1349 Bangladesh
| | - Sabrina Samad Shoily
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Abu Ashfaqur Sajib
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| |
Collapse
|
32
|
Caswell RC, Gunning AC, Owens MM, Ellard S, Wright CF. Assessing the clinical utility of protein structural analysis in genomic variant classification: experiences from a diagnostic laboratory. Genome Med 2022; 14:77. [PMID: 35869530 PMCID: PMC9308257 DOI: 10.1186/s13073-022-01082-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 07/04/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The widespread clinical application of genome-wide sequencing has resulted in many new diagnoses for rare genetic conditions, but testing regularly identifies variants of uncertain significance (VUS). The remarkable rise in the amount of genomic data has been paralleled by a rise in the number of protein structures that are now publicly available, which may have clinical utility for the interpretation of missense and in-frame insertions or deletions. METHODS Within a UK National Health Service genomic medicine diagnostic laboratory, we investigated the number of VUS over a 5-year period that were evaluated using protein structural analysis and how often this analysis aided variant classification. RESULTS We found 99 novel missense and in-frame variants across 67 genes that were initially classified as VUS by our diagnostic laboratory using standard variant classification guidelines and for which further analysis of protein structure was requested. Evidence from protein structural analysis was used in the re-assessment of 64 variants, of which 47 were subsequently reclassified as pathogenic or likely pathogenic and 17 remained as VUS. We identified several case studies where protein structural analysis aided variant interpretation by predicting disease mechanisms that were consistent with the observed phenotypes, including loss-of-function through thermodynamic destabilisation or disruption of ligand binding, and gain-of-function through de-repression or escape from proteasomal degradation. CONCLUSIONS We have shown that using in silico protein structural analysis can aid classification of VUS and give insights into the mechanisms of pathogenicity. Based on our experience, we propose a generic evidence-based workflow for incorporating protein structural information into diagnostic practice to facilitate variant classification.
Collapse
Affiliation(s)
- Richard C Caswell
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, EX2 5DW, UK.
| | - Adam C Gunning
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, EX2 5DW, UK
- Institute of Biomedical and Clinical Science, University of Exeter School of Medicine, Exeter, EX2 5DW, UK
| | - Martina M Owens
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, EX2 5DW, UK
| | - Sian Ellard
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, EX2 5DW, UK
- Institute of Biomedical and Clinical Science, University of Exeter School of Medicine, Exeter, EX2 5DW, UK
| | - Caroline F Wright
- Institute of Biomedical and Clinical Science, University of Exeter School of Medicine, Exeter, EX2 5DW, UK.
| |
Collapse
|
33
|
Gibson JT, Sadeghi-Alavijeh O, Gale DP, Rothe H, Savige J. Pathogenicity of missense variants affecting the collagen IV α5 carboxy non-collagenous domain in X-linked Alport syndrome. Sci Rep 2022; 12:11257. [PMID: 35789182 PMCID: PMC9253329 DOI: 10.1038/s41598-022-14928-x] [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: 02/20/2022] [Accepted: 06/15/2022] [Indexed: 12/05/2022] Open
Abstract
X-linked Alport syndrome is a genetic kidney disease caused by pathogenic COL4A5 variants, but little is known of the consequences of missense variants affecting the NC1 domain of the corresponding collagen IV α5 chain. This study examined these variants in a normal (gnomAD) and other databases (LOVD, Clin Var and 100,000 Genomes Project) to determine their pathogenicity and clinical significance. Males with Cys substitutions in the collagen IV α5 NC1 domain reported in LOVD (n = 25) were examined for typical Alport features, including age at kidney failure. All NC1 variants in LOVD (n = 86) were then assessed for structural damage using an online computational tool, Missense3D. Variants in the ClinVar, gnomAD and 100,000 Genomes Project databases were also examined for structural effects. Predicted damage associated with NC1 substitutions was then correlated with the level of conservation of the affected residues. Cys substitutions in males were associated with the typical features of X-linked Alport syndrome, with a median age at kidney failure of 31 years. NC1 substitutions predicted to cause structural damage were overrepresented in LOVD (p < 0.001), and those affecting Cys residues or 'buried' Gly residues were more common than expected (both p < 0.001). Most NC1 substitutions in gnomAD (88%) were predicted to be structurally-neutral. Substitutions affecting conserved residues resulted in more structural damage than those affecting non-conserved residues (p < 0.001). Many pathogenic missense variants affecting the collagen IV α5 NC1 domain have their effect through molecular structural damage and 3D modelling is a useful tool in their assessment.
Collapse
Affiliation(s)
- Joel T Gibson
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Omid Sadeghi-Alavijeh
- Department of Renal Medicine, University College London, London, UK
- Genomics England, Queen Mary University of London, London, UK
| | - Daniel P Gale
- Department of Renal Medicine, University College London, London, UK
- Genomics England, Queen Mary University of London, London, UK
| | - Hansjörg Rothe
- Centre for Nephrology and Metabolic Disorders, 02943, Weisswasser, Germany
| | - Judy Savige
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Parkville, VIC, 3050, Australia.
- Genomics England, Queen Mary University of London, London, UK.
| |
Collapse
|
34
|
Complex effects on Ca V2.1 channel gating caused by a CACNA1A variant associated with a severe neurodevelopmental disorder. Sci Rep 2022; 12:9186. [PMID: 35655070 PMCID: PMC9163077 DOI: 10.1038/s41598-022-12789-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/16/2022] [Indexed: 01/25/2023] Open
Abstract
P/Q-type Ca2+ currents mediated by CaV2.1 channels are essential for active neurotransmitter release at neuromuscular junctions and many central synapses. Mutations in CACNA1A, the gene encoding the principal CaV2.1 α1A subunit, cause a broad spectrum of neurological disorders. Typically, gain-of-function (GOF) mutations are associated with migraine and epilepsy while loss-of-function (LOF) mutations are causative for episodic and congenital ataxias. However, a cluster of severe CaV2.1 channelopathies have overlapping presentations which suggests that channel dysfunction in these disorders cannot always be defined bimodally as GOF or LOF. In particular, the R1667P mutation causes focal seizures, generalized hypotonia, dysarthria, congenital ataxia and, in one case, cerebral edema leading ultimately to death. Here, we demonstrate that the R1667P mutation causes both channel GOF (hyperpolarizing voltage-dependence of activation, slowed deactivation) and LOF (slowed activation kinetics) when expressed heterologously in tsA-201 cells. We also observed a substantial reduction in Ca2+ current density in this heterologous system. These changes in channel gating and availability/expression manifested in diminished Ca2+ flux during action potential-like stimuli. However, the integrated Ca2+ fluxes were no different when normalized to tail current amplitude measured upon repolarization from the reversal potential. In summary, our findings indicate a complex functional effect of R1667P and support the idea that pathological missense mutations in CaV2.1 may not represent exclusively GOF or LOF.
Collapse
|
35
|
Alburaiky S, Taylor J, O'Grady G, Thomson G, Perry D, England EM, Yap P. Cochlear nerve deficiency in SOX11-related Coffin-Siris syndrome. Am J Med Genet A 2022; 188:2460-2465. [PMID: 35642566 DOI: 10.1002/ajmg.a.62851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 04/01/2022] [Accepted: 04/23/2022] [Indexed: 11/10/2022]
Abstract
The phenotypic spectrum of SOX11-related Coffin-Siris syndrome (CSS) is expanding with reports of new associations. SOX11 is implicated in neurogenesis and inner ear development. Cochlear nerve deficiency, absence or hypoplasia, is commonly associated with cochlear canal stenosis or with CHARGE syndrome, a monogenic condition that affects inner ear development. SOX11 is a transcription factor essential for neuronal identity, highly correlated with the expression of CHD7, which regulates SOX11. We present two unrelated probands, each with novel de novo SOX11 likely pathogenic variants and phenotypic manifestations of CSS including global developmental delay, growth deficiency, and hypoplastic nails. They have unilateral sensorineural hearing loss due to cochlear nerve deficiency confirmed on MRI. SOX11 is implicated in sensory neuron survival and maturation. It is highly expressed in the developing inner ear. Homozygous ablation of SOX11 in a mouse model resulted in a reduction in sensory neuron survival and decreased axonal growth. A heterozygous knockout mice model had hearing impairment with grossly normal inner ear structures like the two probands reported. We propose cochlear nerve deficiency as a new phenotypic feature of SOX11-related CSS. Magnetic resonance imaging is useful in delineating the cochlear nerve deficiency and other CSS-related brain malformations.
Collapse
Affiliation(s)
- Salam Alburaiky
- Genetic Health Service New Zealand-Northern Hub, Auckland, New Zealand
| | - Juliet Taylor
- Genetic Health Service New Zealand-Northern Hub, Auckland, New Zealand
| | - Gina O'Grady
- Department of Paediatric Neurology, Starship Children's Hospital, Auckland, New Zealand
| | - Glen Thomson
- Department of Paediatric Radiology, Starship Children's Hospital, Auckland, New Zealand
| | - David Perry
- Department of Paediatric Radiology, Starship Children's Hospital, Auckland, New Zealand
| | - Eleina M England
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Patrick Yap
- Genetic Health Service New Zealand-Northern Hub, Auckland, New Zealand.,Department of Molecular Medicine and Pathology, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
36
|
Radusky LG, Serrano L. pyFoldX: enabling biomolecular analysis and engineering along structural ensembles. Bioinformatics 2022; 38:2353-2355. [PMID: 35176149 PMCID: PMC9004634 DOI: 10.1093/bioinformatics/btac072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/19/2021] [Accepted: 02/09/2022] [Indexed: 02/03/2023] Open
Abstract
SUMMARY Recent years have seen an increase in the number of structures available, not only for new proteins but also for the same protein crystallized with different molecules and proteins. While protein design software has proven to be successful in designing and modifying proteins, they can also be overly sensitive to small conformational differences between structures of the same protein. To cope with this, we introduce here pyFoldX, a python library that allows the integrative analysis of structures of the same protein using FoldX, an established forcefield and modelling software. The library offers new functionalities for handling different structures of the same protein, an improved molecular parametrization module and an easy integration with the data analysis ecosystem of the python programming language. AVAILABILITY AND IMPLEMENTATION pyFoldX rely on the FoldX software for energy calculations and modelling, which can be downloaded upon registration in http://foldxsuite.crg.eu/ and its licence is free of charge for academics. The pyFoldX library is open-source. Full details on installation, tutorials covering the library functionality and the scripts used to generate the data and figures presented in this paper are available at https://github.com/leandroradusky/pyFoldX. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Leandro G Radusky
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, 08003 Barcelona, Spain
| | | |
Collapse
|
37
|
PDBe-KB consortium, Varadi M, Anyango S, Armstrong D, Berrisford J, Choudhary P, Deshpande M, Nadzirin N, Nair SS, Pravda L, Tanweer A, Al-Lazikani B, Andreini C, Barton GJ, Bednar D, Berka K, Blundell T, Brock KP, Carazo JM, Damborsky J, David A, Dey S, Dunbrack R, Recio JF, Fraternali F, Gibson T, Helmer-Citterich M, Hoksza D, Hopf T, Jakubec D, Kannan N, Krivak R, Kumar M, Levy ED, London N, Macias JR, Srivatsan MM, Marks DS, Martens L, McGowan SA, McGreig JE, Modi V, Parra RG, Pepe G, Piovesan D, Prilusky J, Putignano V, Radusky LG, Ramasamy P, Rausch AO, Reuter N, Rodriguez LA, Rollins NJ, Rosato A, Rubach P, Serrano L, Singh G, Skoda P, Sorzano COS, Stourac J, Sulkowska JI, Svobodova R, Tichshenko N, Tosatto SCE, Vranken W, Wass MN, Xue D, Zaidman D, Thornton J, Sternberg M, Orengo C, Velankar S. PDBe-KB: collaboratively defining the biological context of structural data. Nucleic Acids Res 2022; 50:D534-D542. [PMID: 34755867 PMCID: PMC8728252 DOI: 10.1093/nar/gkab988] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/01/2021] [Accepted: 10/14/2021] [Indexed: 12/15/2022] Open
Abstract
The Protein Data Bank in Europe - Knowledge Base (PDBe-KB, https://pdbe-kb.org) is an open collaboration between world-leading specialist data resources contributing functional and biophysical annotations derived from or relevant to the Protein Data Bank (PDB). The goal of PDBe-KB is to place macromolecular structure data in their biological context by developing standardised data exchange formats and integrating functional annotations from the contributing partner resources into a knowledge graph that can provide valuable biological insights. Since we described PDBe-KB in 2019, there have been significant improvements in the variety of available annotation data sets and user functionality. Here, we provide an overview of the consortium, highlighting the addition of annotations such as predicted covalent binders, phosphorylation sites, effects of mutations on the protein structure and energetic local frustration. In addition, we describe a library of reusable web-based visualisation components and introduce new features such as a bulk download data service and a novel superposition service that generates clusters of superposed protein chains weekly for the whole PDB archive.
Collapse
|
38
|
Tan H, Reed S. Metabolovigilance: Associating Drug Metabolites with Adverse Drug Reactions. Mol Inform 2022; 41:e2100261. [PMID: 34994061 DOI: 10.1002/minf.202100261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 01/03/2022] [Indexed: 11/05/2022]
Abstract
The Metabolovigilance database (https://pharmacogenomics.clas.ucdenver.edu/pharmacogenomics/side-effect/) is a single repository of information on over 15,920 pharmaceuticals and the compounds expected to result from metabolism of these drugs. Metabolovigilance functions as both a web server, providing data directly to users and as a web application, applying user inputs to create logic statements that curate the data presented or downloaded. Using this tool, it is easy to collect information on drugs, their side effects, and the metabolites associated with specific side effects. Information on these compounds can be sorted based on physical properties of the drugs and their metabolites. All of this information can be viewed, sorted, and downloaded for use in other applications. This open-access tool will facilitate molecular studies on the causes of adverse drug reactions and is well suited to integrate with genomic data furthering the goals of personalized medicine.
Collapse
Affiliation(s)
- Henry Tan
- University of Colorado Denver, UNITED STATES
| | - Scott Reed
- University of Colorado Denver, UNITED STATES
| |
Collapse
|
39
|
Yu J, Liao PJ, Xu W, Jones JR, Everman DB, Flanagan-Steet H, Keller TH, Virshup DM. Structural model of human PORCN illuminates disease-associated variants and drug-binding sites. J Cell Sci 2021; 134:273795. [PMID: 34817055 DOI: 10.1242/jcs.259383] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/11/2021] [Indexed: 12/20/2022] Open
Abstract
Wnt signaling is essential for normal development and is a therapeutic target in cancer. The enzyme PORCN, or porcupine, is a membrane-bound O-acyltransferase (MBOAT) that is required for the post-translational modification of all Wnts, adding an essential mono-unsaturated palmitoleic acid to a serine on the tip of Wnt hairpin 2. Inherited mutations in PORCN cause focal dermal hypoplasia, and therapeutic inhibition of PORCN slows the growth of Wnt-dependent cancers. Based on homology to mammalian MBOAT proteins, we developed and validated a structural model of human PORCN. The model accommodates palmitoleoyl-CoA and Wnt hairpin 2 in two tunnels in the conserved catalytic core, shedding light on the catalytic mechanism. The model predicts how previously uncharacterized human variants of uncertain significance can alter PORCN function. Drugs including ETC-159, IWP-L6 and LGK-974 dock in the PORCN catalytic site, providing insights into PORCN pharmacologic inhibition. This structural model enhances our mechanistic understanding of PORCN substrate recognition and catalysis, as well as the inhibition of its enzymatic activity, and can facilitate the development of improved inhibitors and the understanding of disease-relevant PORCN mutants. This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Jia Yu
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857, Singapore
| | - Pei-Ju Liao
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857, Singapore
| | - Weijun Xu
- Discovery Chemistry, Experimental Drug Development Centre, 10 Biopolis Road, Chromos, 138670, Singapore
| | - Julie R Jones
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - David B Everman
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | | | - Thomas H Keller
- Discovery Chemistry, Experimental Drug Development Centre, 10 Biopolis Road, Chromos, 138670, Singapore
| | - David M Virshup
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857, Singapore.,Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
40
|
Comprehensive Study of Human FBXW7 Deleterious nsSNP's Functional Inference and Susceptibility to Gynaecological Cancer. Appl Biochem Biotechnol 2021; 194:407-433. [PMID: 34817806 DOI: 10.1007/s12010-021-03759-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
Cancer is one of the world's major causes of mortality, and it plays a most important role in the world's declining life expectancy. F-box and WD-40 domain protein 7 (FBXW7), a typical participant of the F-box family of proteins, has been considered as an anti-tumor protein and one of the maximum deregulated ubiquitin-proteasome system proteins in uterine carcinosarcoma, endometrial clear cell carcinoma and cervical carcinoma with the greatest prevalence of alterations. FBXW7 variants with known clinical significance, as well as non-synonymous single nucleotide polymorphisms (nsSNPs) in the F-Box and WD40 domains, were evaluated using functionality prediction web resources. Upon analysing the seventy-three deleterious nsSNP's impact on protein stability and function, we identified that forty-one nsSNPs of WD40 domain and three of F-Box domain imply decreased stability of the FBXW7 structure. Next to TP53 and PTEN, FBXW7 was reported with the highest percentage of arginine substitution among mutations related to cancer. The current research concentrated on two arginine residue locations (Arg465, Arg505) within the WD40-repeat domain, which is vital for substrate binding. Computational analysis revealed significant deviation in stability and structural configuration of mutants R505L, R465H, R465P, R505G, R505C, R465C, R505S and R505L structures. Protein-protein interaction network of FBXW7 populated with promising hub proteins NOTCH1, c-Myc, CCNE1, STYX, KLG5, SREB1, NFKB2, SKP1 and CUL1; thus, alteration in the FBXW7 leads to aberration in their signalling pathways as well as their substrate binding ability makes this protein as attractive target for personalized therapeutic intervention.
Collapse
|
41
|
David A, Islam S, Tankhilevich E, Sternberg MJE. The AlphaFold Database of Protein Structures: A Biologist's Guide. J Mol Biol 2021; 434:167336. [PMID: 34757056 PMCID: PMC8783046 DOI: 10.1016/j.jmb.2021.167336] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/06/2023]
Abstract
AlphaFold, the deep learning algorithm developed by DeepMind, recently released the three-dimensional models of the whole human proteome to the scientific community. Here we discuss the advantages, limitations and the still unsolved challenges of the AlphaFold models from the perspective of a biologist, who may not be an expert in structural biology.
Collapse
Affiliation(s)
- Alessia David
- Centre for Integrative System Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK.
| | - Suhail Islam
- Centre for Integrative System Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Evgeny Tankhilevich
- Centre for Integrative System Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Michael J E Sternberg
- Centre for Integrative System Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|