1
|
Sharma D, Dhiman I, Das S, Das DK, Pramanik DD, Dash SK, Pramanik A. Recent Advances in Therapeutic Peptides: Innovations and Applications in Treating Infections and Diseases. ACS OMEGA 2025; 10:17087-17107. [PMID: 40352490 PMCID: PMC12059905 DOI: 10.1021/acsomega.5c02077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025]
Abstract
Peptides have become a powerful frontier in modern medicine, offering a promising therapeutic solution for various diseases and advancing rapidly in pharmaceutical development. These small amino acid chains, with their innovative design, have attracted significant attention due to their versatility and high receptor specificity, which minimizes off-target effects, along with enhanced therapeutic efficacy, biodegradability, low toxicity, and minimal immunogenicity. They are being explored for use in several clinical domains, like metabolic diseases, immunomodulation, and cancer. Furthermore, antimicrobial peptides (AMPs) have grown to be a promising strategy to combat the worldwide challenge of antibiotic resistance, demonstrating promising results against multidrug-resistant organisms. Both natural and engineered peptides have been discovered and investigated, whereas numerous others are progressing toward clinical trials in a number of therapeutic domains. Recent improvements with surface modification, such as peptide engineering, peptide cyclization, PEGylation, and the utilization of synthetic amino acids to enhance their pharmacokinetic profiles and overcome the inherent disadvantages of these peptides have made it possible for the area to continue to advance. Moreover, their therapeutic potential has been further enhanced by innovative delivery methods, such as self-assembling peptides, nanocarriers, and alternate routes of administration. This Review critically states the potential of peptides as versatile therapeutics along with their modifications and advancements to drive the significant progress to treat infections and chronic diseases, along with their potential benefits and challenges.
Collapse
Affiliation(s)
- Deepshikha Sharma
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
| | - Isha Dhiman
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
| | - Swarnali Das
- Department
of Physiology, University of Gour Banga, Malda, West Bengal 732103, India
| | - Deepak Kumar Das
- Department
of Chemistry and Nanoscience, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Devlina Das Pramanik
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
| | - Sandeep Kumar Dash
- Department
of Physiology, University of Gour Banga, Malda, West Bengal 732103, India
| | - Arindam Pramanik
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
- School
of Medicine, University of Leeds, Leeds LS97TF, United Kingdom
| |
Collapse
|
2
|
Moon J. Tandem repeat disorders: from diagnosis to emerging therapeutic strategies. ENCEPHALITIS 2025; 5:27-35. [PMID: 39654412 PMCID: PMC12042693 DOI: 10.47936/encephalitis.2024.00122] [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: 09/15/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 04/18/2025] Open
Abstract
Tandem repeat disorders (TRDs) are genetic conditions characterized by the abnormal expansion of repetitive DNA sequences within specific genes. The growing number of identified TRDs highlights their complexity, with varied molecular mechanisms ranging from toxic protein production and repeat-associated non-AUG translation to RNA toxicity and epigenetic modifications. TRDs also exhibit unique clinical features such as reduced penetrance, anticipation, and repeat motif changes. Advances in molecular diagnostics such as long-read sequencing have significantly improved the detection of TRDs, especially for large or complex repeat expansions. Additionally, emerging therapeutic strategies, particularly antisense oligonucleotides (ASOs) and gene editing technologies, are showing great promise. ASOs in particular have demonstrated success through mechanisms like allele-specific knockdown and splice modulation. In this review, we explore the classification of TRDs, advances in diagnostics, molecular mechanisms, clinical features, and innovative therapeutic strategies, highlighting the need for further research to refine treatments and improve outcomes.
Collapse
Affiliation(s)
- Jangsup Moon
- Rare Disease Center, Department of Genomic Medicine; Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Winarni TI, Aishworiya R, Culpepper H, Zafarullah M, Mendoza G, Wilaisakditipakorn TJ, Likhitweerawong N, Law J, Hagerman R, Tassone F. In Utero Alcohol and Unsuitable Home Environmental Exposure Combined with FMR1 Full Mutation Allele Cause Severe Fragile X Syndrome Phenotypes. Int J Mol Sci 2025; 26:2840. [PMID: 40243429 PMCID: PMC11988866 DOI: 10.3390/ijms26072840] [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: 01/30/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/18/2025] Open
Abstract
We investigated the molecular and clinical profile of five boys carrying the fragile X messenger ribonucleoprotein 1 (FMR1) mutation and who suffered from the effects of prenatal alcohol exposure. Fragile X syndrome (FXS) testing was performed using PCR and Southern Blot analysis, and fragile X messenger ribonucleoprotein protein (FMRP) expression levels were measured by Western blot analysis. Clinical evaluation included cognitive functions, adaptive skills, autism phenotype, and severity of behavior measures. Fetal Alcohol Spectrum Disorder (FASD) was also assessed. Five adopted male siblings were investigated, four of which (cases 1, 2, 3, and 4) were diagnosed with FXS, FASD, and ASD, and one, the fraternal triplet (case 5), was diagnosed with FASD and ASD and no FXS. The molecular profile of case 1 and 2 showed the presence of a hypermethylated full mutation (FM) and the resulting absence of FMRP. Cases 3 and 4 (identical twins) were FM-size mosaics (for the presence of an FM and a deleted allele), resulting in 16% and 50% FMRP expression levels, respectively. FMRP expression level was normal in case 5 (fraternal twin). Severe behavioral problems were observed in all cases, including aggression, tantrum, self-harming, anxiety, and defiant behavior, due to different mutations of the FMR1 gene, in addition to biological exposure, home environmental factors, and potentially to additional background gene effects.
Collapse
Affiliation(s)
- Tri Indah Winarni
- Center for Biomedical Research (CEBIOR), Faculty of Medicine, Universitas Diponegoro, Semarang 50275, Central Java, Indonesia;
| | - Ramkumar Aishworiya
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore 119074, Singapore;
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Hannah Culpepper
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA; (H.C.); (M.Z.); (G.M.)
| | - Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA; (H.C.); (M.Z.); (G.M.)
| | - Guadalupe Mendoza
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA; (H.C.); (M.Z.); (G.M.)
| | - Tanaporn Jasmine Wilaisakditipakorn
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA; (T.J.W.); (J.L.); (R.H.)
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA;
| | - Narueporn Likhitweerawong
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA;
- Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Julie Law
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA; (T.J.W.); (J.L.); (R.H.)
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA;
| | - Randi Hagerman
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA; (T.J.W.); (J.L.); (R.H.)
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA;
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA; (H.C.); (M.Z.); (G.M.)
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA;
| |
Collapse
|
4
|
Chase A, Hamrick L, Arnold H, Smith J, Hantman R, Cortez K, Adayev T, Tortora ND, Dahlman A, Roberts J. Reduced Respiratory Sinus Arrhythmia in Infants with the FMR1 Premutation. Int J Mol Sci 2025; 26:2186. [PMID: 40076819 PMCID: PMC11900448 DOI: 10.3390/ijms26052186] [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: 01/17/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
The fragile X premutation (FXpm) is caused by a CGG repeat expansion on the FMR1 gene. In adults, FXpm is linked with autonomic nervous system (ANS) dysfunction and impairment is associated with CGG repeat length. Given scant infancy research, we examined ANS functioning, via respiratory sinus arrhythmia (RSA) and interbeat interval (IBI), in 82 FXpm and neurotypical infants and their associations with CGG repeats. FXpm infants exhibited lower RSA but no IBI differences. There were no associations between ANS functioning and CGG repeat length. These findings identify an ANS biomarker consistent with the emerging pediatric phenotype in FXpm.
Collapse
Affiliation(s)
- Abigail Chase
- School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| | - Lisa Hamrick
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (L.H.); (H.A.); (J.S.); (R.H.); (K.C.); (A.D.)
- Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC 29208, USA
| | - Holley Arnold
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (L.H.); (H.A.); (J.S.); (R.H.); (K.C.); (A.D.)
- Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC 29208, USA
| | - Jenna Smith
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (L.H.); (H.A.); (J.S.); (R.H.); (K.C.); (A.D.)
- Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC 29208, USA
| | - Rachel Hantman
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (L.H.); (H.A.); (J.S.); (R.H.); (K.C.); (A.D.)
- Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC 29208, USA
| | - Kaitlyn Cortez
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (L.H.); (H.A.); (J.S.); (R.H.); (K.C.); (A.D.)
- Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC 29208, USA
| | - Tatyana Adayev
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (T.A.); (N.D.T.)
| | - Nicole D. Tortora
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (T.A.); (N.D.T.)
| | - Alison Dahlman
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (L.H.); (H.A.); (J.S.); (R.H.); (K.C.); (A.D.)
- Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC 29208, USA
| | - Jane Roberts
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (L.H.); (H.A.); (J.S.); (R.H.); (K.C.); (A.D.)
- Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
5
|
Alshawsh M, Wake M, Gecz J, Corbett M, Saffery R, Pitt J, Greaves R, Williams K, Field M, Cheong J, Bui M, Arora S, Sadedin S, Lunke S, Wall M, Amor DJ, Godler DE. Epigenomic newborn screening for conditions with intellectual disability and autistic features in Australian newborns. Epigenomics 2024; 16:1203-1214. [PMID: 39365098 PMCID: PMC11487350 DOI: 10.1080/17501911.2024.2402681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 09/06/2024] [Indexed: 10/05/2024] Open
Abstract
This study describes a protocol to assess a novel workflow called Epi-Genomic Newborn Screening (EpiGNs) on 100,000 infants from the state of Victoria, Australia. The workflow uses a first-tier screening approach called methylation-specific quantitative melt analysis (MS-QMA), followed by second and third tier testing including targeted methylation and copy number variation analyzes with droplet digital PCR, EpiTYPER system and low-coverage whole genome sequencing. EpiGNs utilizes only two 3.2 mm newborn blood spot punches to screen for genetic conditions, including fragile X syndrome, Prader-Willi syndrome, Angelman syndrome, Dup15q syndrome and sex chromosome aneuploidies. The program aims to: identify clinically actionable methylation screening thresholds for the first-tier screen and estimate prevalence for the conditions screened.
Collapse
Affiliation(s)
- Mohammed Alshawsh
- Department of Paediatrics, Monash University, Melbourne, VIC, 3168, Australia
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
| | - Melissa Wake
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Jozef Gecz
- Robinson Research Institute & Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Mark Corbett
- Robinson Research Institute & Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - James Pitt
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
| | - Ronda Greaves
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Katrina Williams
- Department of Paediatrics, Monash University, Melbourne, VIC, 3168, Australia
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Michael Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, New South Wales, Australia
| | - Jeanie Cheong
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Obstetrics, Gynaecology & Newborn Health, The Royal Women's Hospital, Melbourne, Australia
| | - Minh Bui
- Centre for Epidemiology & Biostatistics, Melbourne School of Population & Global Health, University of Melbourne, Carlton, Australia
| | - Sheena Arora
- Centre for Health Economics Research & Evaluation, University of Technology Sydney, Broadway, NSW, Australia
| | - Simon Sadedin
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Sebastian Lunke
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Meg Wall
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - David J Amor
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - David E Godler
- Murdoch Children's Research Institute, Parkville, VIC, 3052Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
6
|
Li L, Chen R, Zhang H, Li J, Huang H, Weng J, Tan H, Guo T, Wang M, Xie J. The epigenetic modification of DNA methylation in neurological diseases. Front Immunol 2024; 15:1401962. [PMID: 39376563 PMCID: PMC11456496 DOI: 10.3389/fimmu.2024.1401962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
Methylation, a key epigenetic modification, is essential for regulating gene expression and protein function without altering the DNA sequence, contributing to various biological processes, including gene transcription, embryonic development, and cellular functions. Methylation encompasses DNA methylation, RNA methylation and histone modification. Recent research indicates that DNA methylation is vital for establishing and maintaining normal brain functions by modulating the high-order structure of DNA. Alterations in the patterns of DNA methylation can exert significant impacts on both gene expression and cellular function, playing a role in the development of numerous diseases, such as neurological disorders, cardiovascular diseases as well as cancer. Our current understanding of the etiology of neurological diseases emphasizes a multifaceted process that includes neurodegenerative, neuroinflammatory, and neurovascular events. Epigenetic modifications, especially DNA methylation, are fundamental in the control of gene expression and are critical in the onset and progression of neurological disorders. Furthermore, we comprehensively overview the role and mechanism of DNA methylation in in various biological processes and gene regulation in neurological diseases. Understanding the mechanisms and dynamics of DNA methylation in neural development can provide valuable insights into human biology and potentially lead to novel therapies for various neurological diseases.
Collapse
Affiliation(s)
- Linke Li
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Rui Chen
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Stomatology, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Hui Zhang
- Department of Stomatology, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jinsheng Li
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Hao Huang
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jie Weng
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Huan Tan
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Tailin Guo
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Mengyuan Wang
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Stomatology, The Third People’s Hospital of Chengdu and The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Jiang Xie
- Key Laboratory of Drug Targeting and Drug Delivery of Ministry of Education (MOE), Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, West China School of Pharmacy, Sichuan University, Chengdu, China
- Department of Pediatrics, Chengdu Third People’s Hospital, Chengdu, China
| |
Collapse
|
7
|
Zhang B, Zhang J, Chen H, Qiao D, Guo F, Hu X, Qin C, Jin X, Zhang K, Wang C, Cui H, Li S. Role of FMRP in AKT/mTOR pathway-mediated hippocampal autophagy in fragile X syndrome. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111036. [PMID: 38823765 DOI: 10.1016/j.pnpbp.2024.111036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/05/2024] [Accepted: 05/22/2024] [Indexed: 06/03/2024]
Abstract
Fragile X syndrome (FXS) is caused by epigenetic silencing of the Fmr1 gene, leading to the deletion of the coding protein FMRP. FXS induces abnormal hippocampal autophagy and mTOR overactivation. However, it remains unclear whether FMRP regulates hippocampal autophagy through the AKT/mTOR pathway, which influences the neural behavior of FXS. Our study revealed that FMRP deficiency increased the protein levels of p-ULK-1 and p62 and decreased LC3II/LC3I level in Fmr1 knockout (KO) mice. The mouse hippocampal neuronal cell line HT22 with knockdown of Fmr1 by lentivirus showed that the protein levels of p-ULK-1 and p62 were increased, whereas LC3II/LC3I was unchanged. Further observations revealed that FMRP deficiency obstructed autophagic flow in HT22 cells. Therefore, FMRP deficiency inhibited autophagy in the mouse hippocampus and HT22 cells. Moreover, FMRP deficiency increased reactive oxygen species (ROS) level, decreased the co-localization between the mitochondrial outer membrane proteins TOM20 and LC3 in HT22 cells, and caused a decrease in the mitochondrial autophagy protein PINK1 in HT22 cells and Fmr1 KO mice, indicating that FMRP deficiency caused mitochondrial autophagy disorder in HT22 cells and Fmr1 KO mice. To explore the mechanism by which FMRP deficiency inhibits autophagy, we examined the AKT/mTOR signaling pathway in the hippocampus of Fmr1 KO mice, found that FMRP deficiency caused overactivation of the AKT/mTOR pathway. Rapamycin-mediated mTOR inhibition activated and enhanced mitochondrial autophagy. Finally, we examined whether rapamycin affected the neurobehavior of Fmr1 KO mice. The Fmr1 KO mice exhibited stereotypical behavior, impaired social ability, and learning and memory impairment, while rapamycin treatment improved behavioral disorders in Fmr1 KO mice. Thus, our study revealed the molecular mechanism by which FMRP regulates autophagy function, clarifying the role of hippocampal neuron mitochondrial autophagy in the pathogenesis of FXS, and providing novel insights into potential therapeutic targets of FXS.
Collapse
Affiliation(s)
- Bohan Zhang
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China
| | - Jingbao Zhang
- Grade 2020, Basic Medicine, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Huan Chen
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China
| | - Dan Qiao
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China
| | - Fangzhen Guo
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiangting Hu
- Grade 2020, Basic Medicine, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Chao Qin
- Grade 2021, Basic Medicine, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Xiaowen Jin
- Grade 2021, Basic Medicine, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Kaixi Zhang
- Grade 2021, 5+3 Integrated pediatrics, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Chang Wang
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China
| | - Huixian Cui
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China.
| | - Sha Li
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China; The Key Laboratory of Neural and Vascular Biology of Ministry of Education, Shijiazhuang 050017, China.
| |
Collapse
|
8
|
Montanaro FAM, Alfieri P, Caciolo C, Brunetti A, Airoldi A, de Florio A, Tinella L, Bosco A, Vicari S. Fragile X Syndrome and FMR1 premutation: results from a survey on associated conditions and treatment priorities in Italy. Orphanet J Rare Dis 2024; 19:264. [PMID: 38997701 PMCID: PMC11241840 DOI: 10.1186/s13023-024-03272-0] [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: 11/17/2023] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Fragile X Syndrome (FXS) is the most common cause of inherited intellectual disability, caused by CGG-repeat expansions (> 200) in the FMR1 gene leading to lack of expression. Espansion between 55 and 200 triplets fall within the premutation range (PM) and can lead to different clinical conditions, including fragile X- primary ovarian insufficiency (FXPOI), fragile X-associated neuropsychiatric disorders (FXAND) and fragile X-associated tremor/ataxia syndrome (FXTAS). Although there is not a current cure for FXS and for the Fragile X-PM associated conditions (FXPAC), timely diagnosis as well as the implementation of treatment strategies, psychoeducation and behavioral intervention may improve the quality of life (QoL) of people with FXS or FXPAC. With the aim to investigate the main areas of concerns and the priorities of treatment in these populations, the Italian National Fragile X Association in collaboration with Bambino Gesù Children's Hospital, conducted a survey among Italian participants. METHOD Here, we present a survey based on the previous study that Weber and colleagues conducted in 2019 and that aimed to investigate the main symptoms and challenges in American individuals with FXS. The survey has been translated into Italian language to explore FXS needs of treatment also among Italian individuals affected by FXS, family members, caretakers, and professionals. Furthermore, we added a section designated only to people with PM, to investigate the main symptoms, daily living challenges and treatment priorities. RESULTS Anxiety, challenging behaviors, language difficulties and learning disabilities were considered the major areas of concern in FXS, while PM was reported as strongly associated to cognitive problems, social anxiety, and overthinking. Anxiety was reported as a treatment priority in both FXS and PM. CONCLUSION FXS and PM can be associated with a range of cognitive, affective, and physical health complications. Taking a patient-first perspective may help clinicians to better characterize the cognitive-behavioral phenotype associated to these conditions, and eventually to implement tailored therapeutic approaches.
Collapse
Affiliation(s)
- Federica Alice Maria Montanaro
- Child & Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, 00165, Italy
- Department of Education, Psychology, Communication, University of Bari Aldo Moro, Bari, 70122, Italy
| | - Paolo Alfieri
- Child & Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, 00165, Italy.
| | - Cristina Caciolo
- Child & Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, 00165, Italy
| | - Alessia Brunetti
- Associazione Italiana Sindrome X Fragile, Piazza Lima 1, Milan, 20124, Italy
| | - Alessandra Airoldi
- Associazione Italiana Sindrome X Fragile, Piazza Lima 1, Milan, 20124, Italy
| | - Anna de Florio
- Associazione Italiana Sindrome X Fragile, Piazza Lima 1, Milan, 20124, Italy
| | - Luigi Tinella
- Department of Education, Psychology, Communication, University of Bari Aldo Moro, Bari, 70122, Italy
| | - Andrea Bosco
- Department of Education, Psychology, Communication, University of Bari Aldo Moro, Bari, 70122, Italy
| | - Stefano Vicari
- Child & Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, 00165, Italy
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| |
Collapse
|
9
|
Klusek J, Will E, Christensen T, Caravella K, Hogan A, Sun J, Smith J, Fairchild AJ, Roberts JE. Social Communication Delay in an Unbiased Sample of Preschoolers With the FMR1 Premutation. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2024; 67:2316-2332. [PMID: 38889222 PMCID: PMC11253810 DOI: 10.1044/2024_jslhr-23-00580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/20/2023] [Accepted: 04/15/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE The Fragile X Messenger Ribonucleoprotein-1 (FMR1) premutation (FXpm) is a genetic variant that is common in the general population and is associated with health symptoms and disease in adulthood. However, poor understanding of the clinical phenotype during childhood has hindered the development of clinical practice guidelines for screening and intervention. Given that social communication difficulties have been widely documented in adults with the FXpm and are linked with reduced psychosocial functioning, the present study aimed to characterize the communication profile of the FXpm during early childhood. METHOD Eighteen children with the FXpm who were identified through cascade testing (89%) or screening at birth (11%) were compared to 21 matched typically developing children, aged 2-4 years. Participants completed standardized assessments of language (Mullen Scales of Early Learning) and adaptive communication (Vineland Adaptive Behavior Scales-II). Social communication was rated from seminaturalistic interaction samples using the Brief Observation of Social Communication Change. RESULTS Children with the FXpm showed delayed social communication development, with the magnitude of group differences highlighting social communication as a feature that distinguishes children with the FXpm from their peers (p = .046, ηp2 = .12). The groups did not differ on the standardized language and adaptive communication measures (ps > .297, ηp2s < .03). CONCLUSIONS Early screening and treatment of social communication delays may be key to optimizing outcomes for children with the FXpm. Further research is needed to replicate findings in a larger sample, delineate the trajectory and consequences of social communication difficulties across the life span in the FXpm, and determine the potential epidemiological significance of FMR1 as a mediator of developmental communication differences within the general population.
Collapse
Affiliation(s)
- Jessica Klusek
- Department of Communication Sciences and Disorders, Arnold School of Public Health, University of South Carolina, Columbia
| | - Elizabeth Will
- Department of Communication Sciences and Disorders, Arnold School of Public Health, University of South Carolina, Columbia
| | - Thomas Christensen
- Department of Communication Sciences and Disorders, Arnold School of Public Health, University of South Carolina, Columbia
| | - Kelly Caravella
- Department of Psychiatry, Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill School of Medicine
| | - Abigail Hogan
- Department of Communication Sciences and Disorders, Arnold School of Public Health, University of South Carolina, Columbia
| | - Jennifer Sun
- Department of Communication Sciences and Disorders, Arnold School of Public Health, University of South Carolina, Columbia
| | - Jenna Smith
- Department of Psychology, University of South Carolina, Columbia
| | | | - Jane E. Roberts
- Department of Psychology, University of South Carolina, Columbia
| |
Collapse
|
10
|
Tyynismaa H. Parents may prefer prognostic uncertainty about their child's genetic neurodevelopmental condition. Dev Med Child Neurol 2024; 66:828-829. [PMID: 38214948 DOI: 10.1111/dmcn.15853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
This commentary is on the original article by Turbitt et al. on pages 872–881 of this issue.
Collapse
Affiliation(s)
- Henna Tyynismaa
- Department of Medical and Clinical Genetics & Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
11
|
Turbitt E, Bourne M, McEwen A, Amor DJ. Parents' preferences for receiving and discussing prognostic genetic information regarding their children's neurodevelopmental condition: A qualitative study. Dev Med Child Neurol 2024; 66:872-881. [PMID: 38111102 DOI: 10.1111/dmcn.15830] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023]
Abstract
AIM To investigate parents' preferences and motivations for receiving and discussing prognostic genetic test results. METHOD We used a cross-sectional, interpretive description qualitative study design. We collected data through semi-structured interviews with Australian parents, which we analysed using reflexive thematic analysis. RESULTS Parents (n = 32) had a child or children with a genetic neurodevelopmental condition, such as fragile X syndrome, DiGeorge (22q11.2 deletion) syndrome, or Angelman syndrome. Parents of mildly impacted or older children were tolerant to prognostic uncertainty. Parents found conversations about their child's prognosis emotional and preferred to discuss their child's potential strengths and challenges. While most were enthusiastic about prognostic tests and described many motivations for testing, the potential for prognostic information to contribute to a loss of hope and stigmatizing societal views were also discussed. INTERPRETATION Parents had mixed preferences and motivations for acquiring prognostic genetic information about their child, contrasting evidence in other contexts such as cancer where parents typically have minimal tolerance of uncertainty. Health professionals should consider strength-based framing of prognostic information gained from current and emerging technologies when returning results to families. WHAT THIS PAPER ADDS Parents had varied views about receiving prognostic information on their children's neurodevelopmental condition. Some parents preferred prognostic uncertainty about their children's genetic neurodevelopmental condition.
Collapse
Affiliation(s)
- Erin Turbitt
- Discipline of Genetic Counselling, University of Technology Sydney, Ultimo, NSW, Australia
| | - Meg Bourne
- Discipline of Genetic Counselling, University of Technology Sydney, Ultimo, NSW, Australia
| | - Alison McEwen
- Discipline of Genetic Counselling, University of Technology Sydney, Ultimo, NSW, Australia
| | - David J Amor
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
12
|
Csoka AB, El Kouhen N, Bennani S, Getachew B, Aschner M, Tizabi Y. Roles of Epigenetics and Glial Cells in Drug-Induced Autism Spectrum Disorder. Biomolecules 2024; 14:437. [PMID: 38672454 PMCID: PMC11048423 DOI: 10.3390/biom14040437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/31/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by severe deficits in social communication and interaction, repetitive movements, abnormal focusing on objects, or activity that can significantly affect the quality of life of the afflicted. Neuronal and glial cells have been implicated. It has a genetic component but can also be triggered by environmental factors or drugs. For example, prenatal exposure to valproic acid or acetaminophen, or ingestion of propionic acid, can increase the risk of ASD. Recently, epigenetic influences on ASD have come to the forefront of investigations on the etiology, prevention, and treatment of this disorder. Epigenetics refers to DNA modifications that alter gene expression without making any changes to the DNA sequence. Although an increasing number of pharmaceuticals and environmental chemicals are being implicated in the etiology of ASD, here, we specifically focus on the molecular influences of the abovementioned chemicals on epigenetic alterations in neuronal and glial cells and their potential connection to ASD. We conclude that a better understanding of these phenomena can lead to more effective interventions in ASD.
Collapse
Affiliation(s)
- Antonei B. Csoka
- Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA
| | - Nacer El Kouhen
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20100, Morocco
| | - Samia Bennani
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20100, Morocco
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| |
Collapse
|
13
|
Seem K, Kaur S, Kumar S, Mohapatra T. Epigenome editing for targeted DNA (de)methylation: a new perspective in modulating gene expression. Crit Rev Biochem Mol Biol 2024; 59:69-98. [PMID: 38440883 DOI: 10.1080/10409238.2024.2320659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/15/2024] [Indexed: 03/06/2024]
Abstract
Traditionally, it has been believed that inheritance is driven as phenotypic variations resulting from changes in DNA sequence. However, this paradigm has been challenged and redefined in the contemporary era of epigenetics. The changes in DNA methylation, histone modification, non-coding RNA biogenesis, and chromatin remodeling play crucial roles in genomic functions and regulation of gene expression. More importantly, some of these changes are inherited to the next generations as a part of epigenetic memory and play significant roles in gene expression. The sum total of all changes in DNA bases, histone proteins, and ncRNA biogenesis constitutes the epigenome. Continuous progress in deciphering epigenetic regulations and the existence of heritable epigenetic/epiallelic variations associated with trait of interest enables to deploy epigenome editing tools to modulate gene expression. DNA methylation marks can be utilized in epigenome editing for the manipulation of gene expression. Initially, genome/epigenome editing technologies relied on zinc-finger protein or transcriptional activator-like effector protein. However, the discovery of clustered regulatory interspaced short palindromic repeats CRISPR)/deadCRISPR-associated protein 9 (dCas9) enabled epigenome editing to be more specific/efficient for targeted DNA (de)methylation. One of the major concerns has been the off-target effects, wherein epigenome editing may unintentionally modify gene/regulatory element which may cause unintended change/harmful effects. Moreover, epigenome editing of germline cell raises several ethical/safety issues. This review focuses on the recent developments in epigenome editing tools/techniques, technological limitations, and future perspectives of this emerging technology in therapeutics for human diseases as well as plant improvement to achieve sustainable developmental goals.
Collapse
Affiliation(s)
- Karishma Seem
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Simardeep Kaur
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Suresh Kumar
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Trilochan Mohapatra
- Protection of Plant Varieties and Farmers' Rights Authority, New Delhi, India
| |
Collapse
|
14
|
Wu Y, Song T, Xu Q. R-LOOPs on Short Tandem Repeat Expansion Disorders in Neurodegenerative Diseases. Mol Neurobiol 2023; 60:7185-7195. [PMID: 37540313 DOI: 10.1007/s12035-023-03531-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/24/2023] [Indexed: 08/05/2023]
Abstract
Expansions of short tandem repeats (STRs) have been found to be present in more than 50 diseases and have a close connection with neurodegenerative diseases. Transcriptional silencing and R-LOOP formation, RNA-mediated sequestration of RNA-binding proteins (RBPs), gain-of-function (GOF) proteins containing expanded repeats, and repeat-associated non-AUG (RAN) translation of toxic repeat peptides are some potential molecular mechanisms underlying STR expansion disorders. R-LOOP, a byproduct of transcription, is a three-stranded nucleic acid structure with abnormal accumulation that participates in the pathogenesis of STR expansion disorders by inducing DNA damage and genome instability. R-LOOPs can engender a series of DNA damage, such as DNA double-strand breaks (DSBs), single-strand breaks (SSBs), DNA recombination, or mutations in the DNA replication, transcription, or repair processes. In this review, we provide an in-depth discussion of recent advancements in R-LOOP and systematically elaborate on its genetic destabilizing effects in several neurodegenerative diseases. These molecular mechanisms will provide novel targets for drug design and therapeutic upgrading of these devastating diseases.
Collapse
Affiliation(s)
- Yiting Wu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Tingwei Song
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.
| |
Collapse
|
15
|
Elhawary NA, AlJahdali IA, Abumansour IS, Azher ZA, Falemban AH, Madani WM, Alosaimi W, Alghamdi G, Sindi IA. Phenotypic variability to medication management: an update on fragile X syndrome. Hum Genomics 2023; 17:60. [PMID: 37420260 PMCID: PMC10329374 DOI: 10.1186/s40246-023-00507-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023] Open
Abstract
This review discusses the discovery, epidemiology, pathophysiology, genetic etiology, molecular diagnosis, and medication-based management of fragile X syndrome (FXS). It also highlights the syndrome's variable expressivity and common comorbid and overlapping conditions. FXS is an X-linked dominant disorder associated with a wide spectrum of clinical features, including but not limited to intellectual disability, autism spectrum disorder, language deficits, macroorchidism, seizures, and anxiety. Its prevalence in the general population is approximately 1 in 5000-7000 men and 1 in 4000-6000 women worldwide. FXS is associated with the fragile X messenger ribonucleoprotein 1 (FMR1) gene located at locus Xq27.3 and encodes the fragile X messenger ribonucleoprotein (FMRP). Most individuals with FXS have an FMR1 allele with > 200 CGG repeats (full mutation) and hypermethylation of the CpG island proximal to the repeats, which silences the gene's promoter. Some individuals have mosaicism in the size of the CGG repeats or in hypermethylation of the CpG island, both produce some FMRP and give rise to milder cognitive and behavioral deficits than in non-mosaic individuals with FXS. As in several monogenic disorders, modifier genes influence the penetrance of FMR1 mutations and FXS's variable expressivity by regulating the pathophysiological mechanisms related to the syndrome's behavioral features. Although there is no cure for FXS, prenatal molecular diagnostic testing is recommended to facilitate early diagnosis. Pharmacologic agents can reduce some behavioral features of FXS, and researchers are investigating whether gene editing can be used to demethylate the FMR1 promoter region to improve patient outcomes. Moreover, clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 and developed nuclease defective Cas9 (dCas9) strategies have promised options of genome editing in gain-of-function mutations to rewrite new genetic information into a specified DNA site, are also being studied.
Collapse
Affiliation(s)
- Nasser A. Elhawary
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Imad A. AlJahdali
- Department of Community Medicine, College of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Iman S. Abumansour
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Zohor A. Azher
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Alaa H. Falemban
- Department of Pharmacology and Toxicology, College of Medicine, Umm Al-Qura University, Mecca, 24382 Saudi Arabia
| | - Wefaq M. Madani
- Department of Hematology and Immunology, Faculty of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Wafaa Alosaimi
- Department of Hematology, Maternity and Children Hospital, Mecca, Saudi Arabia
| | - Ghydda Alghamdi
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Ikhlas A. Sindi
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
- Preparatory Year Program, Batterjee Medical College, Jeddah, 21442 Saudi Arabia
| |
Collapse
|
16
|
Yano N, Fedulov AV. Targeted DNA Demethylation: Vectors, Effectors and Perspectives. Biomedicines 2023; 11:biomedicines11051334. [PMID: 37239005 DOI: 10.3390/biomedicines11051334] [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: 03/28/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Aberrant DNA hypermethylation at regulatory cis-elements of particular genes is seen in a plethora of pathological conditions including cardiovascular, neurological, immunological, gastrointestinal and renal diseases, as well as in cancer, diabetes and others. Thus, approaches for experimental and therapeutic DNA demethylation have a great potential to demonstrate mechanistic importance, and even causality of epigenetic alterations, and may open novel avenues to epigenetic cures. However, existing methods based on DNA methyltransferase inhibitors that elicit genome-wide demethylation are not suitable for treatment of diseases with specific epimutations and provide a limited experimental value. Therefore, gene-specific epigenetic editing is a critical approach for epigenetic re-activation of silenced genes. Site-specific demethylation can be achieved by utilizing sequence-dependent DNA-binding molecules such as zinc finger protein array (ZFA), transcription activator-like effector (TALE) and clustered regularly interspaced short palindromic repeat-associated dead Cas9 (CRISPR/dCas9). Synthetic proteins, where these DNA-binding domains are fused with the DNA demethylases such as ten-eleven translocation (Tet) and thymine DNA glycosylase (TDG) enzymes, successfully induced or enhanced transcriptional responsiveness at targeted loci. However, a number of challenges, including the dependence on transgenesis for delivery of the fusion constructs, remain issues to be solved. In this review, we detail current and potential approaches to gene-specific DNA demethylation as a novel epigenetic editing-based therapeutic strategy.
Collapse
Affiliation(s)
- Naohiro Yano
- Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Alexey V Fedulov
- Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| |
Collapse
|
17
|
Acero-Garcés DO, Saldarriaga W, Cabal-Herrera AM, Rojas CA, Hagerman RJ. Fragile X Syndrome in children. Colomb Med (Cali) 2023; 54:e4005089. [PMID: 37664646 PMCID: PMC10469670 DOI: 10.25100/cm.v54i2.5089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 11/28/2022] [Accepted: 05/14/2023] [Indexed: 09/05/2023] Open
Abstract
Fragile X syndrome is caused by the expansion of CGG triplets in the FMR1 gene, which generates epigenetic changes that silence its expression. The absence of the protein coded by this gene, FMRP, causes cellular dysfunction, leading to impaired brain development and functional abnormalities. The physical and neurologic manifestations of the disease appear early in life and may suggest the diagnosis. However, it must be confirmed by molecular tests. It affects multiple areas of daily living and greatly burdens the affected individuals and their families. Fragile X syndrome is the most common monogenic cause of intellectual disability and autism spectrum disorder; the diagnosis should be suspected in every patient with neurodevelopmental delay. Early interventions could improve the functional prognosis of patients with Fragile X syndrome, significantly impacting their quality of life and daily functioning. Therefore, healthcare for children with Fragile X syndrome should include a multidisciplinary approach.
Collapse
Affiliation(s)
| | - Wilmar Saldarriaga
- Universidad del Valle, Facultad de Salud, Escuela de Medicina, Cali, Colombia
- Universidad del Valle, Facultad de Salud, Escuela de Ciencias Básicas, Cali, Colombia
| | | | - Christian A. Rojas
- Universidad del Valle, Facultad de Salud, Escuela de Medicina, Cali, Colombia
| | - Randi J. Hagerman
- University of California, Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA, USA
- Davis Medical Center, Sacramento, CA, USA
- University of California, Department of Pediatrics, Davis, CA, USA
| |
Collapse
|
18
|
Baker EK, Arpone M, Bui M, Kraan CM, Ling L, Francis D, Hunter MF, Rogers C, Field MJ, Santa María L, Faundes V, Curotto B, Morales P, Trigo C, Salas I, Alliende AM, Amor DJ, Godler DE. Tissue mosaicism, FMR1 expression and intellectual functioning in males with fragile X syndrome. Am J Med Genet A 2023; 191:357-369. [PMID: 36349505 PMCID: PMC10952635 DOI: 10.1002/ajmg.a.63027] [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: 06/15/2022] [Revised: 09/13/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022]
Abstract
Fragile X syndrome (FXS) is caused by hypermethylation of the FMR1 promoter due to the full mutation expansion (full mutation [FM]: CGG ≥ 200 repeats) and silencing of FMR1. Assessment of mosaicism for active-unmethylated alleles has prognostic utility. This study examined relationships between FMR1 methylation in different tissues with FMR1 messenger ribonucleic acid (mRNA) and intellectual functioning in 87 males with FXS (1.89-43.17 years of age). Methylation sensitive Southern blot (mSB) and Methylation Specific-Quantitative Melt Aanalysis (MS-QMA) were used to examine FMR1 methylation. FMR1 mRNA levels in blood showed strong relationships with FMR1 methylation assessed using MS-QMA in blood (n = 68; R2 = 0.597; p = 1.4 × 10-10 ) and buccal epithelial cells (BEC) (n = 62; R2 = 0.24; p = 0.003), with these measures also showing relationships with intellectual functioning scores (p < 0.01). However, these relationships were not as strong for mSB, with ~40% of males with only FM alleles that were 100% methylated and non-mosaic by mSB, showing methylation mosaicism by MS-QMA. This was confirmed through presence of detectable levels of FMR1 mRNA in blood. In summary, FMR1 methylation levels in blood and BEC examined by MS-QMA were significantly associated with FMR1 mRNA levels and intellectual functioning in males with FXS. These relationships were not as strong for mSB, which underestimated prevalence of mosaicism.
Collapse
Affiliation(s)
- Emma K. Baker
- Diagnosis and Development, Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- School of Psychology and Public HealthLa Trobe UniversityBundooraVictoriaAustralia
| | - Marta Arpone
- Diagnosis and Development, Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- Brain and Mind, Murdoch Children's Research InstituteRoyal Children's HospitalParkvilleVictoriaAustralia
| | - Minh Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global HealthUniversity of MelbourneMelbourneVictoriaAustralia
| | - Claudine M. Kraan
- Diagnosis and Development, Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Ling Ling
- Diagnosis and Development, Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVictoriaAustralia
| | - David Francis
- Victorian Clinical Genetics Services and Murdoch Children's Research InstituteThe Royal Children's HospitalMelbourneVictoriaAustralia
| | - Mathew F. Hunter
- Monash GeneticsMonash HealthClaytonVictoriaAustralia
- Department of PaediatricsMonash UniversityClaytonVictoriaAustralia
| | - Carolyn Rogers
- Genetics of Learning Disability ServiceHunter GeneticsWaratahNew South WalesAustralia
| | - Michael J. Field
- Genetics of Learning Disability ServiceHunter GeneticsWaratahNew South WalesAustralia
| | - Lorena Santa María
- Molecular and Cytogenetics LaboratoryINTA University of ChileSantiagoChile
| | - Víctor Faundes
- Molecular and Cytogenetics LaboratoryINTA University of ChileSantiagoChile
| | - Bianca Curotto
- Molecular and Cytogenetics LaboratoryINTA University of ChileSantiagoChile
| | - Paulina Morales
- Molecular and Cytogenetics LaboratoryINTA University of ChileSantiagoChile
| | - Cesar Trigo
- Molecular and Cytogenetics LaboratoryINTA University of ChileSantiagoChile
| | - Isabel Salas
- Molecular and Cytogenetics LaboratoryINTA University of ChileSantiagoChile
| | | | - David J. Amor
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- Neurodisability and Rehabilitation, Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVictoriaAustralia
| | - David E. Godler
- Diagnosis and Development, Murdoch Children's Research InstituteRoyal Children's HospitalMelbourneVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
19
|
Legüe M. Relevancia de los mecanismos epigenéticos en el neurodesarrollo normal y consecuencias de sus perturbaciones. REVISTA MÉDICA CLÍNICA LAS CONDES 2022. [DOI: 10.1016/j.rmclc.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
20
|
Rodrigues B, Gonçalves A, Sousa V, Maia N, Marques I, Vale-Fernandes E, Santos R, Nogueira AJA, Jorge P. Use of the FMR1 Gene Methylation Status to Assess the X-Chromosome Inactivation Pattern: A Stepwise Analysis. Genes (Basel) 2022; 13:419. [PMID: 35327973 PMCID: PMC8951761 DOI: 10.3390/genes13030419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/23/2022] Open
Abstract
X-chromosome inactivation (XCI) is a developmental process to compensate the imbalance in the dosage of X-chromosomal genes in females. A skewing of the XCI pattern may suggest a carrier status for an X-linked disease or explain the presence of a severe phenotype. In these cases, it is important to determine the XCI pattern, conventionally using the gold standard Human Androgen-Receptor Assay (HUMARA), based on the analysis of the methylation status at a polymorphic CAG region in the first exon of the human androgen receptor gene (AR). The aim of this study was to evaluate whether the methylation status of the fragile mental retardation protein translational regulator gene (FMR1) can provide an XCI pattern similar to that obtained by HUMARA. A set of 48 female carriers of FMR1 gene normal-sized alleles was examined using two assays: HUMARA and a FMR1 methylation PCR (mPCR). Ranges were defined to establish the XCI pattern using the methylation pattern of the FMR1 gene by mPCR. Overall, a 77% concordance of the XCI patterns was obtained between the two assays, which led us to propose a set of key points and a stepwise analysis towards obtaining an accurate result for the XCI pattern and to minimize the underlying pitfalls.
Collapse
Affiliation(s)
- Bárbara Rodrigues
- Molecular Genetics Unit, Centro de Genética Médica Dr. Jacinto Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (B.R.); (A.G.); (V.S.); (N.M.); (I.M.); (R.S.)
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal;
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Ana Gonçalves
- Molecular Genetics Unit, Centro de Genética Médica Dr. Jacinto Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (B.R.); (A.G.); (V.S.); (N.M.); (I.M.); (R.S.)
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal;
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Vanessa Sousa
- Molecular Genetics Unit, Centro de Genética Médica Dr. Jacinto Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (B.R.); (A.G.); (V.S.); (N.M.); (I.M.); (R.S.)
| | - Nuno Maia
- Molecular Genetics Unit, Centro de Genética Médica Dr. Jacinto Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (B.R.); (A.G.); (V.S.); (N.M.); (I.M.); (R.S.)
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal;
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Isabel Marques
- Molecular Genetics Unit, Centro de Genética Médica Dr. Jacinto Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (B.R.); (A.G.); (V.S.); (N.M.); (I.M.); (R.S.)
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal;
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Emídio Vale-Fernandes
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal;
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
- Centre for Medically Assisted Procreation/Public Gamete Bank, Centro Materno-Infantil do Norte Dr. Albino Aroso (CMIN), Centro Hospitalar Universitário do Porto (CHUPorto), 4050-651 Porto, Portugal
| | - Rosário Santos
- Molecular Genetics Unit, Centro de Genética Médica Dr. Jacinto Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (B.R.); (A.G.); (V.S.); (N.M.); (I.M.); (R.S.)
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal;
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - António J. A. Nogueira
- CESAM—Center for Environmental and Marine Studies, Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Paula Jorge
- Molecular Genetics Unit, Centro de Genética Médica Dr. Jacinto Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (B.R.); (A.G.); (V.S.); (N.M.); (I.M.); (R.S.)
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal;
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| |
Collapse
|
21
|
Ruzic D, Djoković N, Srdić-Rajić T, Echeverria C, Nikolic K, Santibanez JF. Targeting Histone Deacetylases: Opportunities for Cancer Treatment and Chemoprevention. Pharmaceutics 2022; 14:pharmaceutics14010209. [PMID: 35057104 PMCID: PMC8778744 DOI: 10.3390/pharmaceutics14010209] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
The dysregulation of gene expression is a critical event involved in all steps of tumorigenesis. Aberrant histone and non-histone acetylation modifications of gene expression due to the abnormal activation of histone deacetylases (HDAC) have been reported in hematologic and solid types of cancer. In this sense, the cancer-associated epigenetic alterations are promising targets for anticancer therapy and chemoprevention. HDAC inhibitors (HDACi) induce histone hyperacetylation within target proteins, altering cell cycle and proliferation, cell differentiation, and the regulation of cell death programs. Over the last three decades, an increasing number of synthetic and naturally derived compounds, such as dietary-derived products, have been demonstrated to act as HDACi and have provided biological and molecular insights with regard to the role of HDAC in cancer. The first part of this review is focused on the biological roles of the Zinc-dependent HDAC family in malignant diseases. Accordingly, the small-molecules and natural products such as HDACi are described in terms of cancer therapy and chemoprevention. Furthermore, structural considerations are included to improve the HDACi selectivity and combinatory potential with other specific targeting agents in bifunctional inhibitors and proteolysis targeting chimeras. Additionally, clinical trials that combine HDACi with current therapies are discussed, which may open new avenues in terms of the feasibility of HDACi’s future clinical applications in precision cancer therapies.
Collapse
Affiliation(s)
- Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.R.); (N.D.); (K.N.)
| | - Nemanja Djoković
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.R.); (N.D.); (K.N.)
| | - Tatjana Srdić-Rajić
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia;
| | - Cesar Echeverria
- Facultad de Medicina, Universidad de Atacama, Copayapu 485, Copiapo 1531772, Chile;
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.R.); (N.D.); (K.N.)
| | - Juan F. Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotica 4, POB 102, 11129 Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370854, Chile
- Correspondence: ; Tel.: +381-11-2685-788; Fax: +381-11-2643-691
| |
Collapse
|
22
|
Klusek J, Fairchild A, Moser C, Mailick MR, Thurman AJ, Abbeduto L. Family history of FXTAS is associated with age-related cognitive-linguistic decline among mothers with the FMR1 premutation. J Neurodev Disord 2022; 14:7. [PMID: 35026985 PMCID: PMC8903682 DOI: 10.1186/s11689-022-09415-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 01/02/2022] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Women who carry a premutation allele of the FMR1 gene are at increased vulnerability to an array of age-related symptoms and disorders, including age-related decline in select cognitive skills. However, the risk factors for age-related decline are poorly understood, including the potential role of family history and genetic factors. In other forms of pathological aging, early decline in syntactic complexity is observed and predicts the later onset of neurodegenerative disease. To shed light on the earliest signs of degeneration, the present study characterized longitudinal changes in the syntactic complexity of women with the FMR1 premutation across midlife, and associations with family history of fragile X-associated tremor/ataxia syndrome (FXTAS) and CGG repeat length. METHODS Forty-five women with the FMR1 premutation aged 35-64 years at study entry participated in 1-5 longitudinal assessments spaced approximately a year apart (130 observations total). All participants were mothers of children with confirmed fragile X syndrome. Language samples were analyzed for syntactic complexity and participants provided information on family history of FXTAS. CGG repeat length was determined via molecular genetic testing. RESULTS Hierarchical linear models indicated that women who reported a family history of FXTAS exhibited faster age-related decline in syntactic complexity than those without a family history, with that difference emerging as the women reached their mid-50 s. CGG repeat length was not a significant predictor of age-related change. CONCLUSIONS Results suggest that women with the FMR1 premutation who have a family history of FXTAS may be at increased risk for neurodegenerative disease, as indicated by age-related loss of syntactic complexity. Thus, family history of FXTAS may represent a personalized risk factor for age-related disease. Follow-up study is needed to determine whether syntactic decline is an early indicator of FXTAS specifically, as opposed to being a more general age-related cognitive decline associated with the FMR1 premutation.
Collapse
Affiliation(s)
- Jessica Klusek
- Department of Communication Sciences and Disorders, Arnold School of Public Health, University of South Carolina, 1705 College Street, SC 29208, Columbia, USA
| | - Amanda Fairchild
- Department of Psychology, University of South Carolina, 1512 Pendleton Street Columbia, Columbia, SC 29208 USA
| | - Carly Moser
- Department of Communication Sciences and Disorders, Arnold School of Public Health, University of South Carolina, 1705 College Street, SC 29208, Columbia, USA
| | - Marsha R. Mailick
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705 USA
| | - Angela John Thurman
- Department of Psychiatry and Behavioral Sciences and MIND Institute, University of California Davis Health, 2825 50th Street, Sacramento, CA 95817 USA
| | - Leonard Abbeduto
- Department of Psychiatry and Behavioral Sciences and MIND Institute, University of California Davis Health, 2825 50th Street, Sacramento, CA 95817 USA
| |
Collapse
|
23
|
Zhang W, Li D, Pang N, Jiang L, Li B, Ye F, He F, Chen S, Liu F, Peng J, Yin J, Yin F. The second-tier status of fragile X syndrome testing for unexplained intellectual disability/global developmental delay in the era of next-generation sequencing. Front Pediatr 2022; 10:911805. [PMID: 35935362 PMCID: PMC9353215 DOI: 10.3389/fped.2022.911805] [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: 04/03/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Although many unexplained intellectual disability/global developmental delay (ID/GDD) individuals have benefited from the excellent detection yield of copy number variations and next-generation sequencing testing, many individuals still who suffer from ID/GDD of unexplained etiology. In this study, we investigated the applicability of fragile X syndrome (FXS) testing in unexplained ID/GDD individuals with negative or absent genetic testing. METHODS In this study, we used the triplet repeat primed polymerase chain reaction to evaluate the value and application of fragile X testing in unexplained ID/GDD individuals with negative or absent genetic testing (n = 681) from three hospitals. RESULTS Of the 681 ID/GDD individuals with negative or absent genetic testing results detected by FXS testing, 12 men and one woman were positive. This corresponded to a diagnostic yield of 1.9% for FXS testing in our cohort. All FXS individuals had either a family history of ID/GDD or suggestive clinical features. The detection yield of FXS testing in ID/GDD individuals who completed genetic testing (2.70%, 12/438) was significantly higher than in individuals without any genetic testing (0.40%, 1/243). CONCLUSIONS This is the first report of FXS testing in ID/GDD individuals who lacked previous genetic testing, which promotes standardization of the FXS diagnostic process. These results highlight the utility of FXS testing of unexplained ID/GDD individuals with negative results from standard genetic testing. In the era of next-generation sequencing, FXS testing is more suitable as a second-tier choice and provides clinicians and geneticists with auxiliary references for tracing the etiology of ID/GDD.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China.,Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Dong Li
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China.,Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Nan Pang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China.,Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Li Jiang
- Department of Neurology, Children's Hospital Affiliated to Chongqing Medical University, Chongqing, China
| | - Baomin Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Fanghua Ye
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China.,Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Shimeng Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China.,Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Fangyun Liu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China.,Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China.,Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Jinghua Yin
- Department of Pathophysiology, Xiangya Hospital, Central South University, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China.,Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Shah M, Selvanathan A, Baynam G, Berman Y, Boughtwood T, Freckmann M, Parasivam G, White SM, Grainger N, Kirk EP, Ma ASL, Sachdev R. Paediatric genomic testing: Navigating genomic reports for the general paediatrician. J Paediatr Child Health 2022; 58:8-15. [PMID: 34427008 PMCID: PMC9292248 DOI: 10.1111/jpc.15703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 11/28/2022]
Abstract
Monogenic rare disorders contribute significantly to paediatric morbidity and mortality, and elucidation of the underlying genetic cause may have benefits for patients, families and clinicians. Advances in genomic technology have enabled diagnostic yields of up to 50% in some paediatric cohorts. This has led to an increase in the uptake of genetic testing across paediatric disciplines. This can place an increased burden on paediatricians, who may now be responsible for interpreting and explaining test results to patients. However, genomic results can be complex, and sometimes inconclusive for the ordering paediatrician. Results may also cause uncertainty and anxiety for patients and their families. The paediatrician's genetic literacy and knowledge of genetic principles are therefore critical to inform discussions with families and guide ongoing patient care. Here, we present four hypothetical case vignettes where genomic testing is undertaken, and discuss possible results and their implications for paediatricians and families. We also provide a list of key terms for paediatricians.
Collapse
Affiliation(s)
- Margit Shah
- Centre for Clinical GeneticsSydney Children's Hospital, Sydney Children's Hospitals NetworkSydneyNew South WalesAustralia,Department of Clinical GeneticsChildren's Hospital at Westmead, Sydney Children's Hospitals NetworkSydneyNew South WalesAustralia,Faculty of Health and Medical ScienceUniversity of SydneySydneyNew South WalesAustralia
| | - Arthavan Selvanathan
- Genetic Metabolic Disorders ServiceSydney Children's Hospitals NetworkSydneyNew South WalesAustralia
| | - Gareth Baynam
- Genetic Services of Western AustraliaKing Edward Memorial HospitalPerthWestern AustraliaAustralia,Western Australian Register of Developmental AnomaliesKing Edward Memorial HospitalPerthWestern AustraliaAustralia
| | - Yemima Berman
- Department of Clinical GeneticsRoyal North Shore HospitalSydneyNew South WalesAustralia,Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
| | - Tiffany Boughtwood
- Australian GenomicsMelbourneVictoriaAustralia,Murdoch Children’s Research InstituteMelbourneVictoriaAustralia
| | - Mary‐Louise Freckmann
- Department of Clinical GeneticsRoyal North Shore HospitalSydneyNew South WalesAustralia,ACT Genetics ServiceThe Canberra HospitalCanberraAustralian Capital TerritoryAustralia
| | - Gayathri Parasivam
- NSW Health Centre for Genetics EducationRoyal North Shore HospitalSydneyNew South WalesAustralia,Present address:
Women's and Children's HospitalAdelaideSouth AustraliaAustralia
| | - Susan M White
- Victorian Clinical Genetics ServicesMelbourneVictoriaAustralia,Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
| | - Natalie Grainger
- NSW Health Centre for Genetics EducationRoyal North Shore HospitalSydneyNew South WalesAustralia
| | - Edwin P Kirk
- Centre for Clinical GeneticsSydney Children's Hospital, Sydney Children's Hospitals NetworkSydneyNew South WalesAustralia,School of Women's and Children's HealthUniversity of New South WalesSydneyNew South WalesAustralia,NSW Health Pathology Randwick Genomics LaboratorySydneyNew South WalesAustralia
| | - Alan SL Ma
- Department of Clinical GeneticsChildren's Hospital at Westmead, Sydney Children's Hospitals NetworkSydneyNew South WalesAustralia,Specialty of Genomic MedicineUniversity of SydneySydneyNew South WalesAustralia
| | - Rani Sachdev
- Centre for Clinical GeneticsSydney Children's Hospital, Sydney Children's Hospitals NetworkSydneyNew South WalesAustralia,School of Women's and Children's HealthUniversity of New South WalesSydneyNew South WalesAustralia
| |
Collapse
|
25
|
|
26
|
Payán-Gómez C, Ramirez-Cheyne J, Saldarriaga W. Variable Expressivity in Fragile X Syndrome: Towards the Identification of Molecular Characteristics That Modify the Phenotype. Appl Clin Genet 2021; 14:305-312. [PMID: 34262328 PMCID: PMC8273740 DOI: 10.2147/tacg.s265835] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/21/2021] [Indexed: 12/18/2022] Open
Abstract
Fragile X syndrome (FXS), is an X-linked inherited genetic disease. FXS is the leading cause of inherited intellectual disability and autism in the world. Those affected are characterized by intellectual disability, language deficit, typical facies, and macroorchidism. Alterations in the FMR1 gene have been associated with FXS. The majority of people with this condition have an allele with an expansion of more than 200 repeats in a tract of CGGs within the 5' untranslated region, and this expansion is associated with a hypermethylated state of the gene promoter. FXS has incomplete penetrance and variable expressivity. Intellectual disability is present in 100% of males and 60% of females. Autism spectrum disorder symptoms appear in 50% to 60% of males and 20% of females. Other characteristics such as behavioral and physical alterations have significant variations in presentation frequency. The molecular causes of the variable phenotype in FXS patients are becoming clear: these causes are related to the FMR1 gene itself and to secondary, modifying gene effects. In FXS patients, size and methylation mosaicisms are common. Secondary to mosaicism, there is a variation in the quantity of FMR1 mRNA and the protein coded by the gene Fragile Mental Retardation Protein (FMRP). Potential modifier genes have also been proposed, with conflicting results. Characterizing patients according to CGG expansion, methylation status, concentration of mRNA and FMRP, and genotypification for possible modifier genes in a clinical setting offers an opportunity to identify predictors for treatment response evaluation. When intervention strategies become available to modulate the course of the disease they could be crucial for selecting patients and identifying the best therapeutic intervention. The purpose of this review is to present the information available about the molecular causes of the variability of the expression incomplete penetrance and variable expressivity in FXS and their potential clinical applications.
Collapse
Affiliation(s)
- César Payán-Gómez
- Deparment of Biology, Faculty of Natural Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Julian Ramirez-Cheyne
- Health Faculty, Universidad del Valle, Cali, Colombia
- Hospital Universitario del Valle, Cali, Colombia
| | - Wilmar Saldarriaga
- Health Faculty, Universidad del Valle, Cali, Colombia
- Hospital Universitario del Valle, Cali, Colombia
| |
Collapse
|
27
|
Detection of Cryptic Fragile X Full Mutation Alleles by Southern Blot in a Female and Her Foetal DNA via Chorionic Villus Sampling, Complicated by Mosaicism for 45,X0/46,XX/47,XXX. Genes (Basel) 2021; 12:genes12060798. [PMID: 34073864 PMCID: PMC8225079 DOI: 10.3390/genes12060798] [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: 04/08/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 11/23/2022] Open
Abstract
We describe a female with a 72 CGG FMR1 premutation (PM) (CGG 55–199) and family history of fragile X syndrome (FXS), referred for prenatal testing. The proband had a high risk of having an affected pregnancy with a full mutation allele (FM) (CGG > 200), that causes FXS through hypermethylation of the FMR1 promoter. The CGG sizing analysis in this study used AmplideX triplet repeat primed polymerase chain reaction (TP-PCR) and long-range methylation sensitive PCR (mPCR). These methods detected a 73 CGG PM allele in the proband’s blood, and a 164 CGG PM allele in her male cultured chorionic villus sample (CVS). In contrast, the Southern blot analysis showed mosaicism for: (i) a PM (71 CGG) and an FM (285–768 CGG) in the proband’s blood, and (ii) a PM (165 CGG) and an FM (408–625 CGG) in the male CVS. The FMR1 methylation analysis, using an EpiTYPER system in the proband, showed levels in the range observed for mosaic Turner syndrome. This was confirmed by molecular and cytogenetic karyotyping, identifying 45,X0/46,XX/47,XXX lines. In conclusion, this case highlights the importance of Southern blot in pre- and postnatal testing for presence of an FM, which was not detected using AmplideX TP-PCR or mPCR in the proband and her CVS.
Collapse
|
28
|
De Jager PL. Deconstructing the epigenomic architecture of human neurodegeneration. Neurobiol Dis 2021; 153:105331. [PMID: 33711493 DOI: 10.1016/j.nbd.2021.105331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 10/21/2022] Open
Abstract
The past 10 years have seen a rapid advance in our ability to profile the epigenome from human pathologic material, opening up new study designs to investigate the role of epigenomic features in human disease. Moderate to large scale studies have now been conducted in the target tissue of neurodegenerative diseases, the brain, and, through the use of rigorous statistical methodologies, have laid a foundation of validated observations and successful study designs that inform our perspective on the role of the epigenome in these diseases, generate new hypotheses, and guide our path forward for a second generation of studies. It is clear that sampling the epigenome is not redundant with other "omic" profiling of the same tissue and that it can serve as an important vehicle for the integration of the effect of multiple environmental exposures on risk of disease. In some cases, change in the epigenome may thus have a causal impact on disease, but we now have evidence that such changes may also mediate some of the effect of tau proteinopathy and that other changes may moderate the impact of genetic risk factors. Thus, the epigenome may be involved at multiple different stages of the sequence of events that leads to human neurodegeneration, and we review the study designs that may begin to guide the development of a more comprehensive perspective on the aging brain's epigenome.
Collapse
Affiliation(s)
- Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, United States of America; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, United States of America; Department of Neurology, Columbia University Medical Center, New York, NY, United States of America.
| |
Collapse
|
29
|
Johnson SJ, Cooper TA. Overlapping mechanisms of lncRNA and expanded microsatellite RNA. WILEY INTERDISCIPLINARY REVIEWS. RNA 2021; 12:e1634. [PMID: 33191580 PMCID: PMC7880542 DOI: 10.1002/wrna.1634] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022]
Abstract
RNA has major regulatory roles in a wide range of biological processes and a surge of RNA research has led to the classification of numerous functional RNA species. One example is long noncoding RNAs (lncRNAs) that are structurally complex transcripts >200 nucleotides (nt) in length and lacking a canonical open reading frame (ORF). Despite a general lack of sequence conservation and low expression levels, many lncRNAs have been shown to have functionality in diverse biological processes as well as in mechanisms of disease. In parallel with the growing understanding of lncRNA functions, there is a growing subset of microsatellite expansion disorders in which the primary mechanism of pathogenesis is an RNA gain of function arising from RNA transcripts from the mutant allele. Microsatellite expansion disorders are caused by an expansion of short (3-10 nt) repeats located within coding genes. Expanded repeat-containing RNA mediates toxicity through multiple mechanisms, the details of which remain only partially understood. The purpose of this review is to highlight the links between functional mechanisms of lncRNAs and the potential pathogenic mechanisms of expanded microsatellite RNA. These shared mechanisms include protein sequestration, peptide translation, micro-RNA (miRNA) processing, and miRNA sequestration. Recognizing the parallels between the normal functions of lncRNAs and the negative impact of expanded microsatellite RNA on biological processes can provide reciprocal understanding to the roles of both RNA species. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Sara J Johnson
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Thomas A Cooper
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
30
|
Kraan CM, Baker EK, Arpone M, Bui M, Ling L, Gamage D, Bretherton L, Rogers C, Field MJ, Wotton TL, Francis D, Hunter MF, Cohen J, Amor DJ, Godler DE. DNA Methylation at Birth Predicts Intellectual Functioning and Autism Features in Children with Fragile X Syndrome. Int J Mol Sci 2020; 21:ijms21207735. [PMID: 33086711 PMCID: PMC7589848 DOI: 10.3390/ijms21207735] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
Fragile X syndrome (FXS) is a leading single-gene cause of intellectual disability (ID) with autism features. This study analysed diagnostic and prognostic utility of the Fragile X-Related Epigenetic Element 2 DNA methylation (FREE2m) assessed by Methylation Specific-Quantitative Melt Analysis and the EpiTYPER system, in retrospectively retrieved newborn blood spots (NBS) and newly created dried blood spots (DBS) from 65 children with FXS (~2–17 years). A further 168 NBS from infants from the general population were used to establish control reference ranges, in both sexes. FREE2m analysis showed sensitivity and specificity approaching 100%. In FXS males, NBS FREE2m strongly correlated with intellectual functioning and autism features, however associations were not as strong for FXS females. Fragile X mental retardation 1 gene (FMR1) mRNA levels in blood were correlated with FREE2m in both NBS and DBS, for both sexes. In females, DNAm was significantly increased at birth with a decrease in childhood. The findings support the use of FREE2m analysis in newborns for screening, diagnostic and prognostic testing in FXS.
Collapse
Affiliation(s)
- Claudine M Kraan
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne VIC 3052, Australia; (C.M.K.); (E.K.B.); (M.A.); (L.L.); (D.G.); (L.B.); (D.J.A.)
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville VIC 3052, Australia
| | - Emma K Baker
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne VIC 3052, Australia; (C.M.K.); (E.K.B.); (M.A.); (L.L.); (D.G.); (L.B.); (D.J.A.)
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville VIC 3052, Australia
- School of Psychology and Public Health, La Trobe University, Bundoora VIC 3086, Australia
| | - Marta Arpone
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne VIC 3052, Australia; (C.M.K.); (E.K.B.); (M.A.); (L.L.); (D.G.); (L.B.); (D.J.A.)
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville VIC 3052, Australia
- Brain and Mind, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville VIC 3052, Australia
| | - Minh Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne VIC 3052, Australia;
| | - Ling Ling
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne VIC 3052, Australia; (C.M.K.); (E.K.B.); (M.A.); (L.L.); (D.G.); (L.B.); (D.J.A.)
| | - Dinusha Gamage
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne VIC 3052, Australia; (C.M.K.); (E.K.B.); (M.A.); (L.L.); (D.G.); (L.B.); (D.J.A.)
| | - Lesley Bretherton
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne VIC 3052, Australia; (C.M.K.); (E.K.B.); (M.A.); (L.L.); (D.G.); (L.B.); (D.J.A.)
| | - Carolyn Rogers
- Genetics of Learning Disability Service (GOLD service), Hunter Genetics, Newcastle NSW 2298, Australia; (C.R.); (M.J.F.)
| | - Michael J Field
- Genetics of Learning Disability Service (GOLD service), Hunter Genetics, Newcastle NSW 2298, Australia; (C.R.); (M.J.F.)
| | - Tiffany L Wotton
- New South Wales Newborn Screening Program, Children’s Hospital at Westmead, Sydney NSW 2145, Australia;
| | - David Francis
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne VIC 3052, Australia;
| | - Matt F Hunter
- Monash Genetics, Monash Health, Clayton, VIC 3168, Australia;
| | - Jonathan Cohen
- Centre for Developmental Disability Health Victoria, Monash University, Doveton VIC 3177, Australia;
- Fragile X Alliance Inc., North Caulfield VIC 3161, Australia
| | - David J Amor
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne VIC 3052, Australia; (C.M.K.); (E.K.B.); (M.A.); (L.L.); (D.G.); (L.B.); (D.J.A.)
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville VIC 3052, Australia
| | - David E Godler
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne VIC 3052, Australia; (C.M.K.); (E.K.B.); (M.A.); (L.L.); (D.G.); (L.B.); (D.J.A.)
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville VIC 3052, Australia
- Correspondence: ; Tel.: +613-8341-6496
| |
Collapse
|
31
|
Lee EC, Hu VW. Phenotypic Subtyping and Re-Analysis of Existing Methylation Data from Autistic Probands in Simplex Families Reveal ASD Subtype-Associated Differentially Methylated Genes and Biological Functions. Int J Mol Sci 2020; 21:E6877. [PMID: 32961747 PMCID: PMC7555936 DOI: 10.3390/ijms21186877] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) describes a group of neurodevelopmental disorders with core deficits in social communication and manifestation of restricted, repetitive, and stereotyped behaviors. Despite the core symptomatology, ASD is extremely heterogeneous with respect to the severity of symptoms and behaviors. This heterogeneity presents an inherent challenge to all large-scale genome-wide omics analyses. In the present study, we address this heterogeneity by stratifying ASD probands from simplex families according to the severity of behavioral scores on the Autism Diagnostic Interview-Revised diagnostic instrument, followed by re-analysis of existing DNA methylation data from individuals in three ASD subphenotypes in comparison to that of their respective unaffected siblings. We demonstrate that subphenotyping of cases enables the identification of over 1.6 times the number of statistically significant differentially methylated regions (DMR) and DMR-associated genes (DAGs) between cases and controls, compared to that identified when all cases are combined. Our analyses also reveal ASD-related neurological functions and comorbidities that are enriched among DAGs in each phenotypic subgroup but not in the combined case group. Moreover, relational gene networks constructed with the DAGs reveal signaling pathways associated with specific functions and comorbidities. In addition, a network comprised of DAGs shared among all ASD subgroups and the combined case group is enriched in genes involved in inflammatory responses, suggesting that neuroinflammation may be a common theme underlying core features of ASD. These findings demonstrate the value of phenotype definition in methylomic analyses of ASD and may aid in the development of subtype-directed diagnostics and therapeutics.
Collapse
Affiliation(s)
| | - Valerie W. Hu
- Department of Biochemistry and Molecular Medicine, The George Washington University, School of Medicine and Health Sciences, Washington, DC 20037, USA;
| |
Collapse
|
32
|
DNA methylation analysis for screening and diagnostic testing in neurodevelopmental disorders. Essays Biochem 2020; 63:785-795. [PMID: 31696914 DOI: 10.1042/ebc20190056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
Abstract
DNA methylation (mDNA) plays an important role in the pathogenesis of neurodevelopmental disorders (NDDs), however its use in diagnostic testing has been largely restricted to a handful of methods for locus-specific analysis in monogenic syndromes. Recent studies employing genome-wide methylation analysis (GWMA) have explored utility of a single array-based test to detect methylation changes in probands negative by exome sequencing, and to diagnose different monogenic NDDs with defined epigenetic signatures. While this may be a more efficient approach, several significant barriers remain. These include non-uniform and low coverage of regulatory regions that may have CG-rich sequences, and lower analytical sensitivity as compared with locus-specific analyses that may result in methylation mosaicism not being detected. A major challenge associated with the above technologies, regardless of whether the analysis is locus specific or genome wide, is the technical bias introduced by indirect analysis of methylation. This review summarizes evidence from the most recent studies in this field and discusses future directions, including direct analysis of methylation using long-read technologies and detection of 5-methylcytosine (5-mC or total mDNA) and 5-hydroxymethylacytosine (5-hmC) as biomarkers of NDDs.
Collapse
|
33
|
Doerfler W. Essential concepts are missing: Foreign DNA in food invades the organisms' cells and can lead to stochastic epigenetic alterations with a wide range of possible pathogenetic consequences. Clin Epigenetics 2020; 12:21. [PMID: 32033622 PMCID: PMC7007663 DOI: 10.1186/s13148-020-0813-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/14/2020] [Indexed: 11/10/2022] Open
Abstract
In this article, a new concept for general pathogenesis has been proposed. Advances in molecular genetics have led to the realization that essential concepts in the framework of molecular biology are still missing. Clinical medicine is plagued by similar shortcomings: The questioning of current paradigms could open new vistas and invite challenging approaches. This article presents an unconventional idea. Foreign DNA which is regularly ingested with the essential food supply is not completely degraded. Small quantities of fragmented DNA rather persist transiently in the gastro-intestinal tract of mice and can be traced to various organ systems, except for cells in the germ line. Foreign DNA entering and persisting in mammalian cells can stochastically lead to genome-wide alterations of transcriptional and CpG DNA methylation profiles. In the course of food-ingested DNA invading somatic cells, completely new cell types can be generated which might be involved in the causation of common ailments. Projects emanating from this perception merit critical analysis and rigorous pursuit.
Collapse
Affiliation(s)
- Walter Doerfler
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany. .,Institute of Genetics, University of Cologne, Cologne, Germany.
| |
Collapse
|
34
|
Baker EK, Arpone M, Vera SA, Bretherton L, Ure A, Kraan CM, Bui M, Ling L, Francis D, Hunter MF, Elliott J, Rogers C, Field MJ, Cohen J, Maria LS, Faundes V, Curotto B, Morales P, Trigo C, Salas I, Alliende AM, Amor DJ, Godler DE. Intellectual functioning and behavioural features associated with mosaicism in fragile X syndrome. J Neurodev Disord 2019; 11:41. [PMID: 31878865 PMCID: PMC6933737 DOI: 10.1186/s11689-019-9288-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Background Fragile X syndrome (FXS) is a common cause of intellectual disability and autism spectrum disorder (ASD) usually associated with a CGG expansion, termed full mutation (FM: CGG ≥ 200), increased DNA methylation of the FMR1 promoter and silencing of the gene. Mosaicism for presence of cells with either methylated FM or smaller unmethylated pre-mutation (PM: CGG 55–199) alleles in the same individual have been associated with better cognitive functioning. This study compares age- and sex-matched FM-only and PM/FM mosaic individuals on intellectual functioning, ASD features and maladaptive behaviours. Methods This study comprised a large international cohort of 126 male and female participants with FXS (aged 1.15 to 43.17 years) separated into FM-only and PM/FM mosaic groups (90 males, 77.8% FM-only; 36 females, 77.8% FM-only). Intellectual functioning was assessed with age appropriate developmental or intelligence tests. The Autism Diagnostic Observation Schedule-2nd Edition was used to examine ASD features while the Aberrant Behavior Checklist-Community assessed maladaptive behaviours. Results Comparing males and females (FM-only + PM/FM mosaic), males had poorer intellectual functioning on all domains (p < 0.0001). Although females had less ASD features and less parent-reported maladaptive behaviours, these differences were no longer significant after controlling for intellectual functioning. Participants with PM/FM mosaicism, regardless of sex, presented with better intellectual functioning and less maladaptive behaviours compared with their age- and sex-matched FM-only counterparts (p < 0.05). ASD features were similar between FM-only and PM/FM mosaics within each sex, after controlling for overall intellectual functioning. Conclusions Males with FXS had significantly lower intellectual functioning than females with FXS. However, there were no significant differences in ASD features and maladaptive behaviours, after controlling for intellectual functioning, independent of the presence or absence of mosaicism. This suggests that interventions that primarily target cognitive abilities may in turn reduce the severity of maladaptive behaviours including ASD features in FXS.
Collapse
Affiliation(s)
- Emma K Baker
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia. .,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia. .,School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia.
| | - Marta Arpone
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,Brain and Mind, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Solange Aliaga Vera
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Lesley Bretherton
- Brain and Mind, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Alexandra Ure
- Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Pediatrics, Monash University, Clayton, VIC, Australia
| | - Claudine M Kraan
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Minh Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Carlton, VIC, Australia
| | - Ling Ling
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - David Francis
- Victorian Clinical Genetics Services and Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Matthew F Hunter
- Department of Pediatrics, Monash University, Clayton, VIC, Australia.,Monash Genetics, Monash Health, Melbourne, VIC, Australia
| | - Justine Elliott
- Victorian Clinical Genetics Services and Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Carolyn Rogers
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - Michael J Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - Jonathan Cohen
- Fragile X Alliance Inc, Centre for Developmental Disability Health Victoria, Monash University, North Caulfield, Clayton, VIC, Australia
| | - Lorena Santa Maria
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Victor Faundes
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Bianca Curotto
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Paulina Morales
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Cesar Trigo
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Isabel Salas
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Angelica M Alliende
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - David J Amor
- Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - David E Godler
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
35
|
Hensel CH, Vanzo RJ, Martin MM, Ling L, Aliaga SM, Bui M, Francis DI, Twede H, Field MH, Morison JW, Amor DJ, Godler DE. Abnormally Methylated FMR1 in Absence of a Detectable Full Mutation in a U.S.A Patient Cohort Referred for Fragile X Testing. Sci Rep 2019; 9:15315. [PMID: 31653898 PMCID: PMC6814816 DOI: 10.1038/s41598-019-51618-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 09/30/2019] [Indexed: 12/11/2022] Open
Abstract
In 2016, Methylation-Specific Quantitative Melt Analysis (MS-QMA) on 3,340 male probands increased diagnostic yield from 1.60% to 1.84% for fragile X syndrome (FXS) using a pooling approach. In this study probands from Lineagen (UT, U.S.A.) of both sexes were screened using MS-QMA without sample pooling. The cohorts included: (i) 279 probands with no FXS full mutation (FM: CGG > 200) detected by AmplideX CGG sizing; (ii) 374 negative and 47 positive controls. MS-QMA sensitivity and specificity in controls approached 100% for both sexes. For male probands with no FM detected by standard testing (n = 189), MS-QMA identified abnormal DNA methylation (mDNA) in 4% normal size (NS: < 44 CGGs), 6% grey zone (CGG 45–54) and 12% premutation (CGG 54–199) alleles. The abnormal mDNA was confirmed by AmplideX methylation sensitive (m)PCR and EpiTYPER tests. In contrast, no abnormal mDNA was detected in 89 males with NS alleles from the general population. For females, 11% of 43 probands with NS alleles by the AmplideX sizing assay had abnormal mDNA by MS-QMA, with FM / NS mosaicism confirmed by AmplideX mPCR. FMR1 MS-QMA analysis can cost-effectively screen probands of both sexes for methylation and FM mosaicism that may be missed by standard testing.
Collapse
Affiliation(s)
| | - Rena J Vanzo
- Lineagen, Inc., Salt Lake City, UT, United States of America
| | - Megan M Martin
- Lineagen, Inc., Salt Lake City, UT, United States of America
| | - Ling Ling
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Solange M Aliaga
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Minh Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - David I Francis
- Victorian Clinical Genetics Services and Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Hope Twede
- Lineagen, Inc., Salt Lake City, UT, United States of America
| | - Michael H Field
- Genetics of Learning Disability Service (GOLD service), Hunter Genetics, Newcastle, NSW, Australia
| | - Jonathon W Morison
- Business Development and Legal Office, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - David J Amor
- Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,Neurodisability and Rehabilitation Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - David E Godler
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia. .,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
36
|
Significantly Elevated FMR1 mRNA and Mosaicism for Methylated Premutation and Full Mutation Alleles in Two Brothers with Autism Features Referred for Fragile X Testing. Int J Mol Sci 2019; 20:ijms20163907. [PMID: 31405222 PMCID: PMC6721168 DOI: 10.3390/ijms20163907] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/04/2019] [Accepted: 08/07/2019] [Indexed: 11/26/2022] Open
Abstract
Although fragile X syndrome (FXS) is caused by a hypermethylated full mutation (FM) expansion with ≥200 cytosine-guanine-guanine (CGG) repeats, and a decrease in FMR1 mRNA and its protein (FMRP), incomplete silencing has been associated with more severe autism features in FXS males. This study reports on brothers (B1 and B2), aged 5 and 2 years, with autistic features and language delay, but a higher non-verbal IQ in comparison to typical FXS. CGG sizing using AmplideX PCR only identified premutation (PM: 55–199 CGGs) alleles in blood. Similarly, follow-up in B1 only revealed PM alleles in saliva and skin fibroblasts; whereas, an FM expansion was detected in both saliva and buccal DNA of B2. While Southern blot analysis of blood detected an unmethylated FM, methylation analysis with a more sensitive methodology showed that B1 had partially methylated PM alleles in blood and fibroblasts, which were completely unmethylated in buccal and saliva cells. In contrast, B2 was partially methylated in all tested tissues. Moreover, both brothers had FMR1 mRNA ~5 fold higher values than those of controls, FXS and PM cohorts. In conclusion, the presence of unmethylated FM and/or PM in both brothers may lead to an overexpression of toxic expanded mRNA in some cells, which may contribute to neurodevelopmental problems, including elevated autism features.
Collapse
|
37
|
Janssens Y, Wynendaele E, Vanden Berghe W, De Spiegeleer B. Peptides as epigenetic modulators: therapeutic implications. Clin Epigenetics 2019; 11:101. [PMID: 31300053 PMCID: PMC6624906 DOI: 10.1186/s13148-019-0700-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/03/2019] [Indexed: 12/13/2022] Open
Abstract
Peptides originating from different sources (endogenous, food derived, environmental, and synthetic) are able to influence different aspects of epigenetic regulation. Endogenous short peptides, resulting from proteolytic cleavage of proteins or upon translation of non-annotated out of frame transcripts, can block DNA methylation and hereby regulate gene expression. Peptides entering the body by digestion of food-related proteins can modulate DNA methylation and/or histone acetylation while environmental peptides, synthesized by bacteria, fungi, and marine sponges, mainly inhibit histone deacetylation. In addition, synthetic peptides that reverse or inhibit different epigenetic modifications of both histones and the DNA can be developed as well. Next to these DNA and histone modifications, peptides can also influence the expression of non-coding RNAs such as lncRNAs and the maturation of miRNAs. Seen the advantages over small molecules, the development of peptide therapeutics is an interesting approach to treat diseases with a strong epigenetic basis like cancer and Alzheimer’s disease. To date, only a limited number of drugs with a proven epigenetic mechanism of action have been approved by the FDA of which two (romidepsin and nesiritide) are peptides. A large knowledge gap concerning epigenetic effects of peptides is present, and this class of molecules deserves more attention in the development as epigenetic modulators. In addition, none of the currently approved peptide drugs are under investigation for their potential effects on epigenetics, hampering drug repositioning of these peptides to other indications with an epigenetic etiology.
Collapse
Affiliation(s)
- Yorick Janssens
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Evelien Wynendaele
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Wim Vanden Berghe
- Protein Science, Proteomics and Epigenetic Signaling (PPES), Department Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Bart De Spiegeleer
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium.
| |
Collapse
|
38
|
Abstract
Increasing numbers of studies implicate abnormal DNA methylation in cancer and many non-malignant diseases. This is consistent with numerous findings about differentiation-associated changes in DNA methylation at promoters, enhancers, gene bodies, and sites that control higher-order chromatin structure. Abnormal increases or decreases in DNA methylation contribute to or are markers for cancer formation and tumour progression. Aberrant DNA methylation is also associated with neurological diseases, immunological diseases, atherosclerosis, and osteoporosis. In this review, I discuss DNA hypermethylation in disease and its interrelationships with normal development as well as proposed mechanisms for the origin of and pathogenic consequences of disease-associated hypermethylation. Disease-linked DNA hypermethylation can help drive oncogenesis partly by its effects on cancer stem cells and by the CpG island methylator phenotype (CIMP); atherosclerosis by disease-related cell transdifferentiation; autoimmune and neurological diseases through abnormal perturbations of cell memory; and diverse age-associated diseases by age-related accumulation of epigenetic alterations.
Collapse
Affiliation(s)
- Melanie Ehrlich
- Tulane Cancer Center and Tulane Center for Bioinformatics and Genomics, Tulane University Health Sciences Center , New Orleans , LA , USA
| |
Collapse
|
39
|
Gomez JA, Beitnere U, Segal DJ. Live-Animal Epigenome Editing: Convergence of Novel Techniques. Trends Genet 2019; 35:527-541. [PMID: 31128888 DOI: 10.1016/j.tig.2019.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022]
Abstract
Epigenome editing refers to the generation of precise chromatin alterations and their effects on gene expression and cell biology. Until recently, much of the efforts in epigenome editing were limited to tissue culture models of disease. However, the convergence of techniques from different fields including mammalian genetics, virology, and CRISPR engineering is advancing epigenome editing into a new era. Researchers are increasingly embracing the use of multicellular model organisms to test the role of specific chromatin alterations in development and disease. The challenge of successful live-animal epigenomic editing will depend on a well-informed foundation of the current methodologies for cell-specific delivery and editing accuracy. Here we review the opportunities for basic research and therapeutic applications.
Collapse
Affiliation(s)
- J Antonio Gomez
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Ulrika Beitnere
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA 95616, USA
| | - David J Segal
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
40
|
Baker EK, Arpone M, Aliaga SM, Bretherton L, Kraan CM, Bui M, Slater HR, Ling L, Francis D, Hunter MF, Elliott J, Rogers C, Field M, Cohen J, Cornish K, Santa Maria L, Faundes V, Curotto B, Morales P, Trigo C, Salas I, Alliende AM, Amor DJ, Godler DE. Incomplete silencing of full mutation alleles in males with fragile X syndrome is associated with autistic features. Mol Autism 2019; 10:21. [PMID: 31073396 PMCID: PMC6499941 DOI: 10.1186/s13229-019-0271-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/03/2019] [Indexed: 11/10/2022] Open
Abstract
Background Fragile X syndrome (FXS) is a common monogenic cause of intellectual disability with autism features. While it is caused by loss of the FMR1 product (FMRP), mosaicism for active and inactive FMR1 alleles, including alleles termed premutation (PM: 55-199 CGGs), is not uncommon. Importantly, both PM and active full mutation (FM: ≥ 200 CGGs) alleles often express elevated levels of mRNA that are thought to be toxic. This study determined if complete FMR1 mRNA silencing from FM alleles and/or levels of FMR1 mRNA (if present) in blood are associated with intellectual functioning and autism features in FXS. Methods The study cohort included 98 participants (70.4% male) with FXS (FM-only and PM/FM mosaic) aged 1-43 years. A control group of 14 females were used to establish control FMR1 mRNA reference range. Intellectual functioning and autism features were assessed using the Mullen Scales of Early Learning or an age-appropriate Wechsler Scale and the Autism Diagnostic Observation Schedule-2nd Edition (ADOS-2), respectively. FMR1 mRNA was analysed in venous blood collected at the time of assessments, using the real-time PCR relative standard curve method. Results Females with FXS had significantly higher levels of FMR1 mRNA (p < 0.001) than males. FMR1 mRNA levels were positively associated with age (p < 0.001), but not with intellectual functioning and autistic features in females. FM-only males (aged < 19 years) expressing FM FMR1 mRNA had significantly higher ADOS calibrated severity scores compared to FM-only males with completely silenced FMR1 (p = 0.011). However, there were no significant differences between these subgroups on intellectual functioning. In contrast, decreased levels of FMR1 mRNA were associated with decreased intellectual functioning in FXS males (p = 0.029), but not autism features, when combined with the PM/FM mosaic group. Conclusion Incomplete silencing of toxic FM RNA may be associated with autistic features, but not intellectual functioning in FXS males. While decreased levels of mRNA may be more predictive of intellectual functioning than autism features. If confirmed in future studies, these findings may have implications for patient stratification, outcome measure development, and design of clinical and pre-clinical trials in FXS.
Collapse
Affiliation(s)
- Emma K. Baker
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Marta Arpone
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
- Brain and Mind, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Australia
| | - Solange M. Aliaga
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
| | - Lesley Bretherton
- Brain and Mind, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Australia
| | - Claudine M. Kraan
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Minh Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Carlton, Australia
| | - Howard R. Slater
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
| | - Ling Ling
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
| | - David Francis
- Victorian Clinical Genetics Services and Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC Australia
| | - Matthew F. Hunter
- Monash Genetics, Monash Health, Melbourne, VIC Australia
- Department of Paediatrics, Monash University, Clayton, VIC Australia
| | - Justine Elliott
- Victorian Clinical Genetics Services and Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC Australia
| | - Carolyn Rogers
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW Australia
| | - Michael Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW Australia
| | - Jonathan Cohen
- Fragile X Alliance Inc, North Caulfield, VIC and Center for Developmental Disability Health Victoria, Monash University, Clayton, Australia
| | - Kim Cornish
- Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Clayton, VIC Australia
| | - Lorena Santa Maria
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Victor Faundes
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Bianca Curotto
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Paulina Morales
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Cesar Trigo
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Isabel Salas
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Angelica M. Alliende
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - David J. Amor
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
- Neurodisability and Rehabilitation, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Australia
| | - David E. Godler
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| |
Collapse
|
41
|
Doerfler W, Weber S, Naumann A. Inheritable epigenetic response towards foreign DNA entry by mammalian host cells: a guardian of genomic stability. Epigenetics 2018; 13:1141-1153. [PMID: 30458693 DOI: 10.1080/15592294.2018.1549463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Apart from its well-documented role in long-term promoter silencing, the genome-wide distribution patterns of ~ 28 million methylated or unmethylated CpG dinucleotides, e. g. in the human genome, is in search of genetic functions. We have set out to study changes in the cellular CpG methylation profile upon introducing foreign DNA into mammalian cells. As stress factors served the genomic integration of foreign (viral or bacterial plasmid) DNA, virus infections or the immortalization of cells with Epstein Barr Virus (EBV). In all instances investigated, alterations in cellular CpG methylation and transcription profiles were observed to different degrees. In the case of adenovirus DNA integration in adenovirus type 12 (Ad12)-transformed hamster cells, the extensive changes in cellular CpG methylation persisted even after the complete loss of all transgenomic Ad12 DNA. Hence, stress-induced alterations in CpG methylation can be inherited independent of the continued presence of the transgenome. Upon virus infections, changes in cellular CpG methylation appear early after infection. In EBV immortalized as compared to control cells, CpG hypermethylation in the far-upstream region of the human FMR1 promoter decreased four-fold. We conclude that in the wake of cellular stress due to foreign DNA entry, preexisting CpG methylation patterns were altered, possibly at specific CpG dinucleotides. Frequently, transcription patterns were also affected. As a working concept, we view CpG methylation profiles in mammalian genomes as a guarding sensor for genomic stability under epigenetic control. As a caveat towards manipulations of cells with foreign DNA, such cells can no longer be considered identical to their un-manipulated counterparts.
Collapse
Affiliation(s)
- Walter Doerfler
- a Institute for Virology , Friedrich-Alexander University Erlangen-Nürnberg , Erlangen , Germany.,b Institute of Genetics , Cologne University , Cologne , Germany
| | - Stefanie Weber
- a Institute for Virology , Friedrich-Alexander University Erlangen-Nürnberg , Erlangen , Germany
| | - Anja Naumann
- a Institute for Virology , Friedrich-Alexander University Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
42
|
Amor DJ. Investigating the child with intellectual disability. J Paediatr Child Health 2018; 54:1154-1158. [PMID: 30294993 DOI: 10.1111/jpc.14202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 12/01/2022]
Abstract
The search for causation is a key component of the assessment of the child with intellectual disability. Historically, a specific diagnosis has been achievable in only a small minority of these children, but over the last decade, this has changed dramatically such that a specific diagnosis is now possible in about half of all children with intellectual disability. This improvement has been driven by major advances in genetic-testing technologies, the most important of which are chromosome microarray and whole exome sequencing. Simultaneously, these technological advances have revealed many new genetic syndromes that had previously escaped clinical recognition, and demonstrated that the majority of severe intellectual disability is caused by pathogenic gene variants that arise de novo in the child. Although access to genomic testing is currently limited, evidence from health economic studies suggests that this testing is most cost effective when performed early in the patient's diagnostic journey.
Collapse
Affiliation(s)
- David J Amor
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|