1
|
Qi Q, Bian J, Li J, Liu K, Yan F, Hou J. Whole-genome transcriptome and DNA methylome analyses reveal molecular abnormalities during the oocyte-to-embryo transition in preimplantation embryos derived from prepubertal lamb oocytes†. Biol Reprod 2025; 112:824-839. [PMID: 40057970 DOI: 10.1093/biolre/ioaf045] [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: 10/13/2024] [Revised: 01/26/2025] [Accepted: 03/04/2025] [Indexed: 05/16/2025] Open
Abstract
The juvenile in vitro embryo transfer technology holds the potential to accelerate livestock breeding. However, its application is limited due to the weak in vitro development of oocytes and embryos from prepubertal lambs. To dissect the regulatory networks of gene expression of sheep embryos and identify the defects in gene expression in prepubertal lamb embryos during the oocyte-to-embryo transition, full-length RNA sequencing and whole-genome bisulfite sequencing based on trace cells were conducted on in vitro-derived embryos generated from adult sheep and prepubertal lamb oocytes. We found that the maternal transcript degradation occurred selectively in adult sheep embryos in multiple waves and was most completed until the morula stage. Major embryonic genome activation was found to occur at the morula stage. By comparing with the patterns of adult embryos, we observed incomplete maternal transcript degradation and abnormal embryonic genome activation in lamb embryos and analyzed their potential molecular mechanisms. Furthermore, we explored dynamic DNA methylation concerning the paternal and maternal genomes during the preimplantation development of sheep embryos, revealing the negative regulatory role of promoter DNA methylation on embryonic genome activation process. Lamb embryos generally displayed higher DNA methylation levels than adults, potentially repressing the embryonic genome activation gene expression, especially the genes associated with ribosomal and mitochondrial organization. We also found abnormalities in the methylation status of imprinted genes in lamb embryos. Our findings advance the understanding of sheep in vitro embryo development and offer insights for improving the juvenile in vitro embryo transfer technology in livestock.
Collapse
Affiliation(s)
- Qi Qi
- State Key Laboratory of Animal Biotech Breeding and College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jiangyue Bian
- State Key Laboratory of Animal Biotech Breeding and College of Biological Sciences, China Agricultural University, Beijing, China
| | - Junjin Li
- State Key Laboratory of Animal Biotech Breeding and College of Biological Sciences, China Agricultural University, Beijing, China
| | - Kexiong Liu
- State Key Laboratory of Animal Biotech Breeding and College of Biological Sciences, China Agricultural University, Beijing, China
| | - Fengxiang Yan
- State Key Laboratory of Animal Biotech Breeding and College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jian Hou
- State Key Laboratory of Animal Biotech Breeding and College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
2
|
O'Leary EM, Bonthuis PJ. Mom genes and dad genes: genomic imprinting in the regulation of social behaviors. Epigenomics 2025:1-19. [PMID: 40249667 DOI: 10.1080/17501911.2025.2491294] [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: 01/08/2025] [Accepted: 03/31/2025] [Indexed: 04/20/2025] Open
Abstract
Genomic imprinting is an epigenetic phenomenon in mammals that affects brain development and behavior. Imprinting involves the regulation of allelic expression for some genes in offspring that depends on whether alleles are inherited from mothers compared to fathers, and is thought to provide parental control over offspring social behavior phenotypes. Imprinted gene expression is prevalent in the mammalian brain, and human imprinted gene mutations are associated with neurodevelopmental disorders and neurodivergent social behavior in Prader-Willi Syndrome, Angelman Syndrome, and autism. Here, we provide a review of the evidence that imprinted genes influence social behaviors across major neurodevelopmental stages in humans and mouse animal models that include parent-infant interactions, juvenile sociability, and adult aggression, dominance, and sexual behavior.
Collapse
Affiliation(s)
- Erin M O'Leary
- Neuroscience Program, University of Illinois, Urbana, IL, USA
| | - Paul J Bonthuis
- Neuroscience Program, University of Illinois, Urbana, IL, USA
- Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Gene Networks in Neural & Development Plasticity Theme at Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
3
|
Montano C, Timp W. Evolution of genome-wide methylation profiling technologies. Genome Res 2025; 35:572-582. [PMID: 40228903 PMCID: PMC12047278 DOI: 10.1101/gr.278407.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
In this mini-review, we explore the advancements in genome-wide DNA methylation profiling, tracing the evolution from traditional methods such as methylation arrays and whole-genome bisulfite sequencing to the cutting-edge single-molecule profiling enabled by long-read sequencing (LRS) technologies. We highlight how LRS is transforming clinical and translational research, particularly by its ability to simultaneously measure genetic and epigenetic information, providing a more comprehensive understanding of complex disease mechanisms. We discuss current challenges and future directions in the field, emphasizing the need for innovative computational tools and robust, reproducible approaches to fully harness the capabilities of LRS in molecular diagnostics.
Collapse
Affiliation(s)
- Carolina Montano
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA;
| |
Collapse
|
4
|
Kagami M, Hara-Isono K, Sasaki A, Amita M. Association between imprinting disorders and assisted reproductive technologies. Epigenomics 2025; 17:397-410. [PMID: 40033833 PMCID: PMC11980493 DOI: 10.1080/17501911.2025.2471269] [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: 04/29/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Aberrant expression of imprinted genes results in imprinting disorders (IDs). Differentially methylated regions (DMRs) reveal parental-origin-specific DNA methylation on CpGs and regulate the expression of the imprinted genes. One etiology of IDs is epimutation (epi-IDs) induced by some error in the establishment or maintenance of methylation imprint during the processes of gametogenesis, fertilization, or early embryonic development. Therefore, it has been a concern that assisted reproductive technologies (ART) increase the risk for the development of IDs, particularly epi-IDs. We review the effects of ART on DNA methylation of the genome, including DMRs in gametes, embryos, and offspring, and the risk of advanced parental age (a confounding factor of ART) and infertility itself for the development of IDs, particularly epi-IDs.
Collapse
Affiliation(s)
- Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kaori Hara-Isono
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Aiko Sasaki
- Division of Obstetrics, Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Mitsuyoshi Amita
- Division of Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
5
|
Besterman AD, Alnor MA, Castaño M, DeLisi LE, Grice DE, Lohoff FW, Middeldorp CM, Müller DJ, Quattrone D, Nurnberger J, Nurmi EL, Ross DA, Soda T, Schulze TG, Trost B, Vilella E, Yap CX, Zai G, Moreno-De-Luca D. Psychiatric Genetics in Clinical Practice: Essential Knowledge for Mental Health Professionals. Am J Psychiatry 2025:appiajp20240295. [PMID: 40134266 DOI: 10.1176/appi.ajp.20240295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
OBJECTIVE The authors provide recommendations on incorporating recent advances in psychiatric genetics into clinical practice for mental health clinicians. METHOD The International Society for Psychiatric Genetics Education Committee met monthly to come to a consensus on priority topics in psychiatric genetics. Topics were then assigned to small teams of subspecialty experts to summarize the current knowledge base and create an illustrative clinical case. Topics included, familial aggregation, common and rare genetic variants, epigenetics, gene-environment interactions, pharmacogenomics, genetic counseling, and ethical and social implications. Each section was reviewed and revised by all committee members and then finalized by the Committee Chair. RESULTS Key findings highlight the importance of understanding the genetic architecture of psychiatric disorders, the potential applications of genetic information in risk assessment, diagnosis, treatment selection, and patient education, as well as the ethical and social considerations surrounding the use of genetic data. The committee emphasizes the need for a nuanced approach that integrates genetic factors with environmental and experiential factors in a holistic model of care. CONCLUSION As psychiatric genetics continues to evolve rapidly, mental health clinicians must stay informed about the latest findings and their clinical implications. Ongoing education, collaboration with genetics professionals, and effective communication strategies are crucial to harness the power of genetics while avoiding potential pitfalls such as genetic determinism and stigma. The committee recommends a balanced perspective that recognizes the complex interplay of genetic and non-genetic factors in shaping mental health outcomes.
Collapse
Affiliation(s)
- Aaron D Besterman
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Mohamed A Alnor
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Mauricio Castaño
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Lynn E DeLisi
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Dorothy E Grice
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Falk W Lohoff
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Christel M Middeldorp
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Daniel J Müller
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Diego Quattrone
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - John Nurnberger
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Erika L Nurmi
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - David A Ross
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Takahiro Soda
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Thomas G Schulze
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Brett Trost
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Elisabet Vilella
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Chloe X Yap
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Gwyneth Zai
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| | - Daniel Moreno-De-Luca
- University of California San Diego Department of Psychiatry, Rady Children's Hospital San Diego, and Rady Children's Institute for Genomic Medicine, San Diego (Besterman); School of Medicine, University of Khartoum, Khartoum, Sudan and American Center for Psychiatry and Neurology, UAE (Alnor); Department of Mental Health and Human Behaviour, University of Caldas, Manizales, Caldas, Colombia (Castaño); Cambridge Health Alliance and Harvard Medical School, Cambridge, MA (DeLisi); Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York (Grice); National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (Lohoff); Department of Child and Youth Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Amsterdam Public Health Research Institute, The Netherlands (Middeldorp); Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health, Toronto, Canada and Department of Psychiatry, University of Toronto, Canada (Müller); Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, U.K., and Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Psychiatry Section, University of Palermo, Palermo, Italy (Quattrone); Departments of Psychiatry and Medical and Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis (Nurnberger); Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (Nurmi); Department of Psychiatry, University of Alberta Faculty of Medicine and Dentistry, Edmonton (Ross); Center for Autism and Neurodevelopment, Department of Psychiatry, University of Florida College of Medicine, Gainesville (Soda); Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Germany (Schulze); Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada (Trost); Hospital Universitari Institut Pere Mata, Institut d'Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, CIBERSAM-Instituto de Salud Carlos III. Reus, Spain (Vilella); Department of Psychiatry, University of Oxford, and Mater Research Institute, Faculty of Medicine, University of Queensland and Metro South Addiction and Mental Health Service, Brisbane, Australia (Yap); Neurogenetics Section, Molecular Brain Sciences Department, Centre for Addiction and Mental Health, Department of Psychiatry and Institute of Medical Science, University of Toronto, Ontario, Canada (Zai); Precision Medicine in Autism (PRISMA) Group, Division of Child and Adolescent Psychiatry, Department of Psychiatry, Faculty of Medicine and Dentistry; Women and Children's Health Research Institute; Neuroscience and Mental Health Institute; University of Alberta; Alberta Health Services; CASA Mental Health; Edmonton, AB, Canada (Moreno-De-Luca)
| |
Collapse
|
6
|
Fischer LA, Meyer B, Reyes M, Zemke JE, Harrison JK, Park KM, Wang T, Jüppner H, Dietmann S, Theunissen TW. Tracking and mitigating imprint erasure during induction of naive human pluripotency at single-cell resolution. Stem Cell Reports 2025; 20:102419. [PMID: 39952244 PMCID: PMC11960550 DOI: 10.1016/j.stemcr.2025.102419] [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: 06/19/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 02/17/2025] Open
Abstract
Naive human pluripotent stem cells (hPSCs) model the pre-implantation epiblast. However, parent-specific epigenetic marks (imprints) are eroded in naive hPSCs, which represents an important deviation from the epiblast in vivo. To track the dynamics of imprint erasure during naive resetting in real time, we established a dual-colored fluorescent reporter at both alleles of the imprinted SNRPN locus. During primed-to-naive resetting, SNRPN expression becomes biallelic in most naive cells, and biallelic SNRPN expression is irreversible upon re-priming. We utilized this live-cell reporter to evaluate chemical and genetic strategies to minimize imprint erasure. Decreasing the level of MEK/ERK inhibition or overexpressing the KRAB zinc-finger protein ZFP57 protected a subset of imprints during naive resetting. Combining these two strategies protected imprint levels to a further extent than either strategy alone. This study offers an experimental tool to track and enhance imprint stability during transitions between human pluripotent states in vitro.
Collapse
Affiliation(s)
- Laura A Fischer
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brittany Meyer
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Monica Reyes
- Endocrine Unit, Department of Medicine and Pediatric Nephrology Unit, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joseph E Zemke
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jessica K Harrison
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyoung-Mi Park
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ting Wang
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Harald Jüppner
- Endocrine Unit, Department of Medicine and Pediatric Nephrology Unit, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sabine Dietmann
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA; Institute for Informatics (I(2)), Washington University School of Medicine, St. Louis, MO, USA
| | - Thorold W Theunissen
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Rosenski J, Peretz A, Magenheim J, Loyfer N, Shemer R, Glaser B, Dor Y, Kaplan T. Atlas of imprinted and allele-specific DNA methylation in the human body. Nat Commun 2025; 16:2141. [PMID: 40069157 PMCID: PMC11897249 DOI: 10.1038/s41467-025-57433-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Allele-specific DNA methylation reflects genetic variation and parentally-inherited changes, and is involved in gene regulation and pathologies. Yet, our knowledge of this phenomenon is largely limited to blood. Here we present a comprehensive atlas of allele-specific DNA methylation using deep whole-genome sequencing across 39 normal human cell types. We identified 325k regions, covering 6% of the genome and 11% of CpGs, that show a bimodal distribution of methylated and unmethylated molecules. In 34k of these regions, genetic variations at individual alleles segregate with methylation patterns, validating allele-specific methylation. We also identified 460 regions showing parental allele-specific methylation, the majority of which are novel, as well as 78 regions associated with known imprinted genes. Surprisingly, sequence-dependent and parental allele-dependent methylation is often restricted to specific cell types, revealing unappreciated variation of allele-specific methylation across the human body. Finally, we validate tissue-specific, maternal allele-specific methylation of CHD7, offering a potential mechanism for the paternal bias in the inheritance mode of CHARGE syndrome associated with this gene. The atlas provides a resource for studying allele-specific methylation and regulatory mechanisms underlying imprinted expression in specific human cell types.
Collapse
Affiliation(s)
- Jonathan Rosenski
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ayelet Peretz
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Judith Magenheim
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Netanel Loyfer
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ruth Shemer
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Benjamin Glaser
- Department of Endocrinology and Metabolism, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Center for Computational Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Tommy Kaplan
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Center for Computational Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
8
|
Kou S, Lu Z, Deng D, Ye M, Sui Y, Qin L, Feng T, Jiang Z, Meng J, Lin CP, Li X, Liu C, Tang J, Zhang H. Activation of Imprinted Gene PW1 Promotes Cardiac Fibrosis After Ischemic Injury. Circulation 2025; 151:623-639. [PMID: 39704066 DOI: 10.1161/circulationaha.124.070738] [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: 06/06/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Cardiac fibrosis, characterized by excessive extracellular matrix (ECM) deposition in the myocardium, is an important target for heart disease treatments. Pw1 (paternally expressed gene 3) is an imprinted gene expressed from the paternal allele, and de novo purine biosynthesis (DNPB) is a crucial pathway for nucleotide synthesis. However, the roles of PW1 and DNPB in ECM production by cardiac fibroblasts during myocardial ischemia are not yet understood. METHODS To induce myocardial damage, we performed left anterior descending coronary artery ligation. We generated Pw1CreER-2A-eGFP and Pw12A-CreER knock-in mouse lines to evaluate the expression of the 2 Pw1 alleles in normal and injured hearts. Bisulfite sequencing was used to analyze the DNA methylation of the Pw1 imprinting control region. We identified the phosphoribosylformylglycinamidine synthase (Pfas) gene, encoding the DNPB enzyme PFAS, as a direct target of PW1 using chromatin immunoprecipitation sequencing and real-time quantitative polymerase chain reaction. The role of DNPB in ECM production and cardiac fibrosis after injury was examined in vitro using cultured cardiac fibroblasts and in vivo with Pfas-deficient mice. RESULTS Our study demonstrates that myocardial infarction reduces DNA methylation at the imprinting control region of the maternally imprinted gene Pw1, triggering a switch from monoallelic imprinting to biallelic expression of Pw1 in cardiac fibroblasts. In activated cardiac fibroblasts, increased Pw1 expression promotes purine biosynthesis and induces ECM production by transcriptionally activating the DNPB factor Pfas. We identified that DNPB is essential for ECM production in activated fibroblasts and that loss of Pfas in fibroblasts limits cardiac fibrosis and improves heart function after injury. CONCLUSIONS This study demonstrates that Pw1 imprinting is disrupted after injury and reveals a novel role for the downstream target PFAS in ECM production and cardiac fibrogenesis. Targeting the PW1/PFAS signaling pathway presents a promising therapeutic strategy for improving cardiac repair after injury.
Collapse
Affiliation(s)
- Shan Kou
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Science and Technology, Tongji University, Shanghai, China (S.K., Z.L., J.T.)
| | - Zhengkai Lu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Science and Technology, Tongji University, Shanghai, China (S.K., Z.L., J.T.)
| | - Defang Deng
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
| | - Min Ye
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
| | - Yu Sui
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
| | - Lieyang Qin
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (L.Q., C.L.)
| | - Teng Feng
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
| | - Zhen Jiang
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
| | - Jufeng Meng
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
| | - Chao-Po Lin
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
| | - Xiajun Li
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
| | - Chen Liu
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (L.Q., C.L.)
| | - Juan Tang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Science and Technology, Tongji University, Shanghai, China (S.K., Z.L., J.T.)
| | - Hui Zhang
- School of Life Science and Technology (S.K., D.D., M.Y., Y.S., T.F., Z.J., J.M., C.L., X.L., H.Z.), ShanghaiTech University, Shanghai, China
- State Key Laboratory of Advanced Medical Materials and Devices & Shanghai Clinical Research and Trial Center (H.Z.), ShanghaiTech University, Shanghai, China
| |
Collapse
|
9
|
Devinsky O, Coller J, Ahrens-Nicklas R, Liu XS, Ahituv N, Davidson BL, Bishop KM, Weiss Y, Mingorance A. Gene therapies for neurogenetic disorders. Trends Mol Med 2025:S1471-4914(25)00015-2. [PMID: 39966070 DOI: 10.1016/j.molmed.2025.01.015] [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: 11/18/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 02/20/2025]
Abstract
Pathogenic variants in over 1700 genes can cause neurogenetic disorders. Monogenetic diseases are ideal targets for genetic therapies; however, the blood-brain barrier (BBB), post-mitotic neurons, and inefficient delivery platforms make gene therapies for neurogenetic diseases challenging. Following nusinersen's 2016 approval, the development of gene therapies for neurogenetic disorders has advanced rapidly, with new delivery vehicles [e.g., BBB-crossing capsids, engineered viral-like proteins, lipid nanoparticles (LNPs)] and novel therapeutic strategies (e.g., regulatory elements, novel RNA therapeutics, tRNA therapies, epigenetic and gene editing). Patient-led disease foundations have accelerated treatment development by addressing trial readiness and supporting translational research. We review the current landscape and future directions in developing gene therapies for neurogenetic disorders.
Collapse
Affiliation(s)
- Orrin Devinsky
- Department of Neurology, NYU Langone Medical Center, New York, NY, USA; Grossman School of Medicine, New York University, New York, NY, USA.
| | - Jeff Coller
- Department of Molecular Biology and Genetics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Biology, Johns Hopkins University, Baltimore, MD, USA; Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Rebecca Ahrens-Nicklas
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - X Shawn Liu
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Beverly L Davidson
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Yael Weiss
- Mahzi Therapeutics, San Francisco, CA, USA
| | - Ana Mingorance
- Dracaena Consulting, Madrid, Spain; Loulou Foundation, London, UK; CureSHANK, Beverly Hills, CA, USA.
| |
Collapse
|
10
|
Iwasaki Y, Reyes M, Jüppner H, Bastepe M. A biallelically active embryonic enhancer dictates GNAS imprinting through allele-specific conformations. Nat Commun 2025; 16:1377. [PMID: 39910084 PMCID: PMC11799514 DOI: 10.1038/s41467-025-56608-0] [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: 04/30/2024] [Accepted: 01/23/2025] [Indexed: 02/07/2025] Open
Abstract
Genomic imprinting controls parental allele-specific gene expression via epigenetic mechanisms. Abnormal imprinting at the GNAS gene causes multiple phenotypes, including pseudohypoparathyroidism type-1B (PHP1B), a disorder of multihormone resistance. Microdeletions affecting the neighboring STX16 gene ablate an imprinting control region (STX16-ICR) of GNAS and lead to PHP1B upon maternal but not paternal inheritance. Mechanisms behind this imprinted inheritance mode remain unknown. Here, we show that the STX16-ICR forms different chromatin conformations with each GNAS parental allele and enhances two GNAS promoters in human embryonic stem cells. When these cells differentiate toward proximal renal tubule cells, STX16-ICR loses its effect, accompanied by a transition to a somatic cell-specific GNAS imprinting status. The activity of STX16-ICR depends on an OCT4 motif, whose disruption impacts transcript levels differentially on each allele. Therefore, a biallelically active embryonic enhancer dictates GNAS imprinting via different chromatin conformations, underlying the allele-specific pathogenicity of STX16-ICR microdeletions.
Collapse
Affiliation(s)
- Yorihiro Iwasaki
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Monica Reyes
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Harald Jüppner
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Murat Bastepe
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Choza JI, Virani M, Kuhn NC, Adams M, Kochmanski J, Bernstein AI. Parkinson's disease-associated alterations in DNA methylation and hydroxymethylation in human brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.21.595193. [PMID: 39975085 PMCID: PMC11838189 DOI: 10.1101/2024.05.21.595193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Epigenetic mechanisms are mediators of interactions between aging, genetics, and environmental factors in sporadic Parkinson's disease (PD). Multiple studies have explored the DNA modifications in PD, but few focus on 5-hydroxymethylcytosine (5-hmC), which is important in the central nervous system and sensitive to environmental exposures. To date, studies have not differentiated between 5-methylcytosine (5-mC) and 5-hmC or have analyzed them separately. In this study, we modeled paired 5-mC and 5-hmC data simultaneously. We identified 108 cytosines with significant PD-associated shifts between these marks in an enriched neuronal population from PD postmortem parietal cortex, within 83 genes and 34 enhancers associated with 67 genes. These data potentially link epigenetic regulation of genes related to LRRK2 and endolysosomal sort (RAB32 and AGAP1), and genes involved in neuroinflammation, the inflammasome, and neurodevelopment with early changes in PD and suggest that there are significant shifts between 5mC and 5hmC associated with PD in genes not captured by standard methods.
Collapse
Affiliation(s)
- Juliana I Choza
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
| | - Mahek Virani
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
| | - Nathan C Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Marie Adams
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI
| | - Joseph Kochmanski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Alison I Bernstein
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| |
Collapse
|
12
|
Lomberk G, Urrutia R. The triple code model for advancing research in rare and undiagnosed diseases beyond the base pairs. Epigenomics 2025; 17:115-124. [PMID: 39630027 PMCID: PMC11792834 DOI: 10.1080/17501911.2024.2436837] [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: 05/29/2024] [Accepted: 11/26/2024] [Indexed: 02/01/2025] Open
Abstract
Rare and undiagnosed diseases pose significant challenges for understanding their mechanisms, diagnosis, and treatment. The Triple Code Model, an integrative paradigm described here, considers the combined influence of the genetic code, epigenetic code, and nuclear structure (an emerging code), as fundamental biochemical mechanisms underlying many rare diseases. Studies demonstrate dysfunctional membrane and cytoplasmic signals instruct the epigenome to ultimately impact the 3D structure and dynamics of the nucleus, highlighting their close interrelationships. Consequently, this model offers a holistic perspective on rare and undiagnosed diseases by moving beyond a solely genetic view. We propose that this integrated framework will efficiently guide rare disease research by taking it 'Beyond the Base Pairs,' leading to improved diagnostics and personalized treatments.
Collapse
Affiliation(s)
- Gwen Lomberk
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Raul Urrutia
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
13
|
Bellingham M, Evans NP, Lea RG, Padmanabhan V, Sinclair KD. Reproductive and Metabolic Health Following Exposure to Environmental Chemicals: Mechanistic Insights from Mammalian Models. Annu Rev Anim Biosci 2025; 13:411-440. [PMID: 39531389 DOI: 10.1146/annurev-animal-111523-102259] [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] [Indexed: 11/16/2024]
Abstract
The decline in human reproductive and metabolic health over the past 50 years is associated with exposure to complex mixtures of anthropogenic environmental chemicals (ECs). Real-life EC exposure has varied over time and differs across geographical locations. Health-related issues include declining sperm quality, advanced puberty onset, premature ovarian insufficiency, cancer, obesity, and metabolic syndrome. Prospective animal studies with individual and limited EC mixtures support these observations and provide a means to investigate underlying physiological and molecular mechanisms. The greatest impacts of EC exposure are through programming of the developing embryo and/or fetus, with additional placental effects reported in eutherian mammals. Single-chemical effects and mechanistic studies, including transgenerational epigenetic inheritance, have been undertaken in rodents. Important translational models of human exposure are provided by companion animals, due to a shared environment, and sheep exposed to anthropogenic chemical mixtures present in pastures treated with sewage sludge (biosolids). Future animal research should prioritize EC mixtures that extend beyond a single developmental stage and/or generation. This would provide a more representative platform to investigate genetic and underlying mechanisms that explain sexually dimorphic and individual effects that could facilitate mitigation strategies.
Collapse
Affiliation(s)
- Michelle Bellingham
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom;
| | - Neil P Evans
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom;
| | - Richard G Lea
- University of Nottingham, Loughborough, United Kingdom
| | | | | |
Collapse
|
14
|
Ma Y, Yan M, Xie Z, Zhang H, Li Z, Li Y, Yang S, Zhang M, Li W, Li J. Generation of live mice from haploid ESCs with germline-DMR deletions or switch. Cell Discov 2025; 11:5. [PMID: 39833184 PMCID: PMC11747502 DOI: 10.1038/s41421-024-00757-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/01/2024] [Indexed: 01/22/2025] Open
Abstract
Genomic imprinting is required for sexual reproduction and embryonic development of mammals, in which, differentially methylated regions (DMRs) regulate the parent-specific monoallelic expression of imprinted genes. Numerous studies on imprinted genes have highlighted their critical roles in development. However, what imprinting network is essential for development is still unclear. Here, we establish a stepwise system to reconstruct a development-related imprinting network, in which diploid embryonic stem cells (ESCs) are derived by fusing between parthenogenetic (PG)- and androgenetic (AG)-haploid embryonic stem cells (haESCs) with different DMR deletions (termed Ha-Ha-fusion system), followed by tetraploid complementation to produce all-haESC fetuses. Diploid ESCs fused between PG-haESCs carrying 8 maternally-derived DMR deletions and AG-haESCs with 2 paternally-derived DMR deletions give rise to live pups efficiently, among which, one lives to weaning. Strikingly, diploid ESCs derived from the fusion of PG-haESCs with 7 maternal DMR deletions and AG-haESCs with 2 paternal DMR deletions and maternal Snrpn-DMR deletion also support full-term embryonic development. Moreover, embryos reconstructed by injection of AG-haESCs with hypomethylated H19-DMR into oocytes with H19-DMR deletion develop into live mice sustaining inverted allelic gene expression. Together, our findings indicate that restoration of monoallelic expression of 10 imprinted regions is adequate for the full-term development of all-haESC pups, and it works irrespective of their parental origins. Meanwhile, Ha-Ha-fusion system provides a useful tool for deciphering imprinting regulation networks during embryonic development.
Collapse
Affiliation(s)
- Yongjian Ma
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Center for Reproductive Medicine & Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Yan
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenfei Xie
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hongling Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhoujie Li
- Animal Core Facility, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyuan Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Suming Yang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Meiling Zhang
- Center for Reproductive Medicine & Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Li
- Center for Reproductive Medicine & Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jinsong Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
15
|
Geysens M, Huremagic B, Souche E, Breckpot J, Devriendt K, Peeters H, Van Buggenhout G, Van Esch H, Van Den Bogaert K, Vermeesch JR. Clinical evaluation of long-read sequencing-based episignature detection in developmental disorders. Genome Med 2025; 17:1. [PMID: 39789644 PMCID: PMC11721592 DOI: 10.1186/s13073-024-01419-z] [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: 04/22/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND A subset of developmental disorders (DD) is characterized by disease-specific genome-wide methylation changes. These episignatures inform on the underlying pathogenic mechanisms and can be used to assess the pathogenicity of genomic variants as well as confirm clinical diagnoses. Currently, the detection of these episignature requires the use of indirect methylation profiling methodologies. We hypothesized that long-read whole genome sequencing would not only enable the detection of single nucleotide variants and structural variants but also episignatures. METHODS Genome-wide nanopore sequencing was performed in 40 controls and 20 patients with confirmed or suspected episignature-associated DD, representing 13 distinct diseases. Following genomic variant and methylome calling, hierarchical clustering and dimensional reduction were used to determine the compatibility with microarray-based episignatures. Subsequently, we developed a support vector machine (SVM) for the detection of each DD. RESULTS Nanopore sequencing-based methylome patterns were concordant with microarray-based episignatures. Our SVM-based classifier identified the episignatures in 17/19 patients with a (likely) pathogenic variant and none of the controls. The remaining patients in which no episignature was identified were also classified as controls by a commercial microarray assay. In addition, we identified all underlying pathogenic single nucleotide and structural variants and showed haplotype-aware skewed X-inactivation evaluation directs clinical interpretation. CONCLUSION This proof-of-concept study demonstrates nanopore sequencing enables episignature detection. In addition, concurrent haplotyped genomic and epigenomic analyses leverage simultaneous detection of single nucleotide/structural variants, X-inactivation, and imprinting, consolidating a multi-step sequential process into a single diagnostic assay.
Collapse
Affiliation(s)
- Mathilde Geysens
- Laboratory of Cytogenetics and Genome Research, Centre for Human Genetics, KU Leuven, Leuven, 3000, Belgium
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Benjamin Huremagic
- Laboratory of Cytogenetics and Genome Research, Centre for Human Genetics, KU Leuven, Leuven, 3000, Belgium
| | - Erika Souche
- Laboratory of Cytogenetics and Genome Research, Centre for Human Genetics, KU Leuven, Leuven, 3000, Belgium
| | - Jeroen Breckpot
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Koenraad Devriendt
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Hilde Peeters
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Griet Van Buggenhout
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Hilde Van Esch
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Kris Van Den Bogaert
- Laboratory of Cytogenetics and Genome Research, Centre for Human Genetics, KU Leuven, Leuven, 3000, Belgium
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Joris Robert Vermeesch
- Laboratory of Cytogenetics and Genome Research, Centre for Human Genetics, KU Leuven, Leuven, 3000, Belgium.
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium.
| |
Collapse
|
16
|
Dionne O, Sabatié S, Laurent B. Deciphering the physiopathology of neurodevelopmental disorders using brain organoids. Brain 2025; 148:12-26. [PMID: 39222411 PMCID: PMC11706293 DOI: 10.1093/brain/awae281] [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: 03/30/2024] [Revised: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Neurodevelopmental disorders (NDD) encompass a range of conditions marked by abnormal brain development in conjunction with impaired cognitive, emotional and behavioural functions. Transgenic animal models, mainly rodents, traditionally served as key tools for deciphering the molecular mechanisms driving NDD physiopathology and significantly contributed to the development of pharmacological interventions aimed at treating these disorders. However, the efficacy of these treatments in humans has proven to be limited, due in part to the intrinsic constraint of animal models to recapitulate the complex development and structure of the human brain but also to the phenotypic heterogeneity found between affected individuals. Significant advancements in the field of induced pluripotent stem cells (iPSCs) offer a promising avenue for overcoming these challenges. Indeed, the development of advanced differentiation protocols for generating iPSC-derived brain organoids gives an unprecedented opportunity to explore human neurodevelopment. This review provides an overview of how 3D brain organoids have been used to investigate various NDD (i.e. Fragile X syndrome, Rett syndrome, Angelman syndrome, microlissencephaly, Prader-Willi syndrome, Timothy syndrome, tuberous sclerosis syndrome) and elucidate their pathophysiology. We also discuss the benefits and limitations of employing such innovative 3D models compared to animal models and 2D cell culture systems in the realm of personalized medicine.
Collapse
Affiliation(s)
- Olivier Dionne
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Salomé Sabatié
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Benoit Laurent
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5H4, Canada
| |
Collapse
|
17
|
Xu Z, Shi J, Chen Q, Yang S, Wang Z, Xiao B, Lai Z, Jing Y, Li Y, Li X. Regulation of de novo and maintenance DNA methylation by DNA methyltransferases in postimplantation embryos. J Biol Chem 2025; 301:107990. [PMID: 39542247 PMCID: PMC11742614 DOI: 10.1016/j.jbc.2024.107990] [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: 08/25/2024] [Revised: 10/23/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
DNA methylation is mainly catalyzed by three DNA methyltransferase (DNMT) proteins in mammals. Usually DNMT1 is considered the primary DNMT for maintenance DNA methylation, whereas DNMT3A and DNMT3B function in de novo DNA methylation. Interestingly, we found DNMT3A and DNMT3B exerted maintenance and de novo DNA methylation in postimplantation mouse embryos. Together with DNMT1, they maintained DNA methylation at some pluripotent genes and lineage marker genes. Germline-derived DNA methylation at the imprinting control regions (ICRs) is stably maintained in embryos. DNMT1 maintained DNA methylation at most ICRs in postimplantation embryos. Surprisingly, DNA methylation was increased at five ICRs after implantation, and two DNMT3 proteins maintained the newly acquired DNA methylation at two of these five ICRs. Intriguingly, DNMT3A and DNMT3B maintained preexisting DNA methylation at four other ICRs, similar to what we found in embryonic stem cells before. These results suggest that DNA methylation is more dynamic than originally thought during embryogenesis including the ICRs of the imprinted regions. DNMT3A and DNMT3B exert both de novo and maintenance DNA methylation functions after implantation. They maintain large portions of newly acquired DNA methylation at variable degrees across the genome in mouse embryos, together with DNMT1. Furthermore, they contribute to maintenance of preexisting DNA methylation at a subset of ICRs as well as in the CpG islands and certain lineage marker gene. These findings may have some implications for the important roles of DNMT proteins in development and human diseases.
Collapse
Affiliation(s)
- Zhen Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jiajia Shi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Qian Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shuting Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zilin Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Biao Xiao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhijian Lai
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yumeng Jing
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yilin Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiajun Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
18
|
Fabiani M, Margiotti K, Libotte F, Alì C, Zangheri C, Barone MA, Viola A, Cima A, Buonomo G, Cristofano D, Lippa A, Pasquale C, Montanino Oliva M, Mesoraca A, Giorlandino C. Uniparental disomy (UPD) exclusion in embryos following Preimplantation Genetic Testing for Structural Rearrangements (PGT-SR). J Assist Reprod Genet 2025; 42:265-273. [PMID: 39693035 PMCID: PMC11805737 DOI: 10.1007/s10815-024-03352-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024] Open
Abstract
PURPOSE Uniparental disomy (UPD) is a genetic condition which both copies of a chromosome are inherited from a single parent, potentially leading to imprinting disorders. This study aimed to assess the integration of Short Tandem Repeat (STR) analysis into Preimplantation Genetic Testing for Structural Rearrangements (PGT-SR) to assess UPD risk and its impact on selecting euploid embryos for embryo transfer in couples with chromosomal translocations involving imprinted chromosomes. METHODS This study evaluated three couples carrying balanced chromosomal translocations: 45,XX,der(13;14)(q10;q10), 46,XX,t(10;11)(q22;q13), and 45,XY,der(14;15)(q10;q10). STR analysis was performed on trophectoderm (TE) biopsies after Whole Genome Amplification (WGA) after PGT-SR analysis using parental blood samples to assess UPD risk in euploid embryos. Haplotyping was conducted with five to six STR markers specific to each rearranged chromosome to detect UPD in euploid embryos. RESULTS Of the four embryos analyzed across the three families, two couples had euploid embryos that tested negative for UPD. These embryos were successfully transferred, resulting in the birth of two healthy children. In the third family, the euploid embryo also tested negative for UPD but failed to implant after transfer, resulting in no pregnancy. DISCUSSION Despite its rarity, UPD involving imprinted chromosomes poses significant clinical risks, as seen in disorders such as Prader-Willi syndrome and Angelman syndrome. This study highlights the importance of integrating UPD screening into PGT-SR protocols, to detect both heterodisomic and isodisomic UPD events minimizing the risk of severe genetic disorders. CONCLUSION Integrating STR-based UPD screening within PGT-SR workflows is a reliable and cost-effective strategy that enhances embryo selection and mitigates the risk of imprinting disorders. This approach improves reproductive outcomes for families with chromosomal rearrangements, offering a practical advancement in assisted reproduction.
Collapse
Affiliation(s)
- Marco Fabiani
- Department of Human Genetic, Altamedica, Rome, Italy.
| | | | | | - Chiara Alì
- Department of Human Genetic, Altamedica, Rome, Italy
| | | | | | | | | | - Giovanna Buonomo
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | - Danilo Cristofano
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | - Assunta Lippa
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | - Chiara Pasquale
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | - Mario Montanino Oliva
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| | | | - Claudio Giorlandino
- Department of Human Genetic, Altamedica, Rome, Italy
- Department of Reproductive Medicine, Altamedica, Fetal-Maternal Medical Centre, Rome, Italy
| |
Collapse
|
19
|
Sinclair KD. Developmental epigenetics: Understanding genetic and sexually dimorphic responses to parental diet and outcomes following assisted reproduction. J Dairy Sci 2024:S0022-0302(24)01392-4. [PMID: 39701526 DOI: 10.3168/jds.2024-25811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/17/2024] [Indexed: 12/21/2024]
Abstract
The developmental integrity and wellbeing of offspring are influenced by events that occur in utero, particularly around the time of conception. While extraneous factors such as environmental temperature and exposure to environmental chemicals can each have a bearing on these events, the epigenetic mechanisms that direct cellular differentiation during early development in ruminants are best described for studies which have investigated the effects of parental nutrition or pregnancy outcomes following assisted reproduction. In this article the case is made that the genetic constitution of an individual directs epigenetic responses to environmental stimuli, and consideration in this regard is also given to the origins of sexual dimorphism and mechanisms of germline intergenerational inheritance. These aspects are considered in the context of epigenetic modifications that take place during the normal course of gametogenesis and embryogenesis, and again following either dietary or procedural interventions such as embryo culture. A recurring feature of such interventions, irrespective of species, is that one carbon metabolic pathways are invariably disrupted, and this affects the provision of methyl groups for chromatin and RNA methylation. Inter-specific variation in how these pathways operate, both within the liver and in germ cells, indicates that ruminants may be particularly sensitive in this regard. Recent advances in genomic technologies should enable rapid progress in these areas. Knowledge gained can be integrated into breed improvement programs and used to tailor management practices to specific breeds and strains (including sexes) within breeds. Ultimately, consideration should be given to integrating metagenomics into analyses of genetic-directed epigenetic programming of animal development.
Collapse
Affiliation(s)
- Kevin D Sinclair
- School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire, UK, LE12 5RD.
| |
Collapse
|
20
|
Manero-Azua A, Vado Y, Gonzàlez Morlà J, Mogas E, Pereda A, Perez de Nanclares G. Heterodisomy in the GNAS locus is also a cause of pseudohypoparathyroidism type 1B (iPPSD3). Front Endocrinol (Lausanne) 2024; 15:1505244. [PMID: 39736869 PMCID: PMC11682883 DOI: 10.3389/fendo.2024.1505244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/18/2024] [Indexed: 01/01/2025] Open
Abstract
Objective To identify the genetic cause underlying the methylation defect in a patient with clinical suspicion of PHP1B/iPPSD3. Design Imprinting is an epigenetic mechanism that allows the regulation of gene expression. The GNAS locus is one of the loci within the genome that is imprinted. When the methylation pattern is affected, it causes pseudohypoparathyroidism type 1B (PHP1B) or inactivating PTH/PTHrP signaling disorder 3 (iPPSD3). Paternal uniparental isodisomy (iUPDpat) of the chromosomal region comprising the GNAS locus has been described as one of the possible underlying genetic causes of the methylation alteration. Methods We present the case of a patient clinically diagnosed with iPPSD3. We performed a commercial methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA), single-nucleotide polymorphism (SNP) array, and microsatellite study. In addition, we designed a custom MS-MLPA to analyze GNAS and nearby differentially methylated regions (DMRs). Results A methylation defect at the four GNAS-DMRs was detected, confirming the clinical diagnosis. Complementary techniques revealed the presence of a mixed isodisomy and heterodisomy of chromosome 20. Surprisingly, the GNAS locus was located on the heterodisomic zone. Conclusions Paternal uniparental heterodisomy (hUPD) at the GNAS locus is also a genetic defect associated with iPPSD3. In the absence of parental samples, our custom MS-MLPA allows for the detection of a methylation defect at the GNAS locus and flanking DMRs, suggestive of uniparental disomy (UPD). We also suggest updating the actual guidelines to include hUPD at the GNAS locus as a cause of iPPSD3.
Collapse
Affiliation(s)
- Africa Manero-Azua
- Rare Disease Research Group, Molecular (Epi) Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital, Vitoria-Gasteiz, Spain
| | - Yerai Vado
- Rare Disease Research Group, Molecular (Epi) Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital, Vitoria-Gasteiz, Spain
| | | | - Eduard Mogas
- Pediatric Endocrinology Section, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Arrate Pereda
- Rare Disease Research Group, Molecular (Epi) Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital, Vitoria-Gasteiz, Spain
| | - Guiomar Perez de Nanclares
- Rare Disease Research Group, Molecular (Epi) Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital, Vitoria-Gasteiz, Spain
| |
Collapse
|
21
|
Zhang Y, Zheng Y, Yu W, Yang L, Zhang C, Li S, Li S. ZFAT (isoform-specific) and its antisense RNA 1 (ZFAT-AS1) are two allele-specific monoallelically expressed genes in cattle. Anim Genet 2024; 55:820-824. [PMID: 39231103 DOI: 10.1111/age.13473] [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: 04/15/2024] [Accepted: 08/08/2024] [Indexed: 09/06/2024]
Abstract
In mammals, imprinted genes are characterised by a monoallelic expression, which is based on parental origin and is essential for both foetal and placental development. The ZFAT gene encodes a transcriptional factor, and its non-coding antisense RNA, ZFAT-AS1, overlaps with the ZFAT locus. Both ZFAT and ZFAT-AS1 are maternally imprinted in human placentas. In bovines, the imprinting status of the ZFAT and ZFAT-AS1 genes has yet to be reported. In this study, we analysed the allelic expression of three transcript variants (X1-X3) of the bovine ZFAT and ZFAT-AS1 genes in somatic tissues and placentas using a single nucleotide polymorphism-based method. The results showed that bovine ZFAT exhibited isoform-specific paternal expression. The ZFAT X2 variant exhibited monoallelic expression in the bovine placentas and biallelic expression in the six bovine somatic tissues (heart, liver, spleen, lung, kidney and brain). However, the ZFAT X1 and X3 variants were biallelically expressed in both bovine tissues and placentas. A 311 bp bovine ZFAT-AS1 complementary DNA (cDNA) sequence was obtained by aligning the human ZFAT-AS1 cDNA sequence with the bovine genome and conducting reverse transcription polymerase chain reaction amplification. Bovine ZFAT-AS1 have monoallelic expression in bovine placentas and somatic tissues. In addition, the DNA methylation of two regions was characterised, including the partial promoter, and exon 1 and intron 1 regions of ZFAT, and there were no differentially methylated regions.
Collapse
Affiliation(s)
- Yinjiao Zhang
- College of Life Science, Agricultural University of Hebei, Baoding, Hebei, China
| | - Yunchang Zheng
- College of Life Science, Agricultural University of Hebei, Baoding, Hebei, China
| | - Wenli Yu
- Hebei Cattle Industry Technology Research Institute, Shijiazhuang, China
| | - Lidan Yang
- College of Life Science, Agricultural University of Hebei, Baoding, Hebei, China
| | - Cui Zhang
- College of Life Science, Agricultural University of Hebei, Baoding, Hebei, China
| | - Shujing Li
- Hebei Provincial Dairy Cow Breeding Engineering Technology Research Center, Shijiazhuang, China
| | - Shijie Li
- College of Life Science, Agricultural University of Hebei, Baoding, Hebei, China
| |
Collapse
|
22
|
da Silva Santos R, Pascoalino Pinheiro D, Gustavo Hirth C, Barbosa Bezerra MJ, Joyce de Lima Silva-Fernandes I, Andréa da Silva Oliveira F, Viana de Holanda Barros M, Silveira Ramos E, A. Moura A, Filho ODMM, Pessoa C, Miranda Furtado CL. Hypomethylation at H19DMR in penile squamous cell carcinoma is not related to HPV infection. Epigenetics 2024; 19:2305081. [PMID: 38245880 PMCID: PMC10802203 DOI: 10.1080/15592294.2024.2305081] [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: 08/09/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Penile squamous cell carcinoma (SCC) is a rare and aggressive tumour mainly related to lifestyle behaviour and human papillomavirus (HPV) infection. Environmentally induced loss of imprinting (LOI) at the H19 differentially methylated region (H19DMR) is associated with many cancers in the early events of tumorigenesis and may be involved in the pathogenesis of penile SCC. We sought to evaluate the DNA methylation pattern at H19DMR and its association with HPV infection in men with penile SCC by bisulfite sequencing (bis-seq). We observed an average methylation of 32.2% ± 11.6% at the H19DMR of penile SCC and did not observe an association between the p16INK4a+ (p = 0.59) and high-risk HPV+ (p = 0.338) markers with methylation level. The average methylation did not change according to HPV positive for p16INK4a+ or hrHPV+ (35.4% ± 10%) and negative for both markers (32.4% ± 10.1%) groups. As the region analysed has a binding site for the CTCF protein, the hypomethylation at the surrounding CpG sites might alter its insulator function. In addition, there was a positive correlation between intense polymorphonuclear cell infiltration and hypomethylation at H19DMR (p = 0.035). Here, we report that hypomethylation at H19DMR in penile SCC might contribute to tumour progression and aggressiveness regardless of HPV infection.
Collapse
Affiliation(s)
- Renan da Silva Santos
- Department of Physiology and Pharmacology, Drug Research and Development Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | | | | | | | | | - Maisa Viana de Holanda Barros
- Postgraduate Program in Translational Medicine, Drug Research and Development Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Ester Silveira Ramos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Arlindo A. Moura
- Department of Physiology and Pharmacology, Drug Research and Development Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
- Department of Animal Science, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Odorico de Moraes Manoel Filho
- Department of Physiology and Pharmacology, Drug Research and Development Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
- Postgraduate Program in Translational Medicine, Drug Research and Development Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Claudia Pessoa
- Department of Physiology and Pharmacology, Drug Research and Development Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Cristiana Libardi Miranda Furtado
- Postgraduate Program in Translational Medicine, Drug Research and Development Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
- Experimental Biology Center, University of Fortaleza, Fortaleza, Ceará, Brazil
- Graduate Program in Medical Sciences, Universidade de Fortaleza, Fortaleza, Ceará, Brazil
| |
Collapse
|
23
|
Zhou J, Mao R, Gao L, Wang M, Long R, Wang X, Li Z, Jin L, Zhu L. Novel variants in PADI6 genes cause female infertility due to early embryo arrest. J Assist Reprod Genet 2024; 41:3327-3336. [PMID: 39644447 PMCID: PMC11707103 DOI: 10.1007/s10815-024-03332-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/15/2024] [Indexed: 12/09/2024] Open
Abstract
PURPOSE Early embryo arrest is characterized by premature termination of development in preimplantation embryos. Human subcortical maternal complex (SCMC) is a protein complex that is specifically expressed in mammalian oocytes and early embryos and is essential for embryonic cell division. Peptidyl arginine deiminase 6 (PADI6) is proven to be a member of SCMC. Variants in the PADI6 gene have been shown to induce early embryo arrest. In this study, we performed genetic analysis in patients with female infertility due to early embryo arrest to identify the disease-causing gene variants. METHODS Whole-exome sequencing and Sanger sequencing were used to identify the variants in the patients and their families. Western blotting and immunofluorescence staining were used to check the effects of the variants on expression and function of PADI6. RESULTS We identified a novel homozygous variant (c.358A > C [p.Thr120Pro]) and novel compound-heterozygous variants (c.2044C > T [p.Arg682Trp] and c.707dupT [p.Leu237Alafs*24]) in PADI6 in two infertile individuals with early embryo arrest. We found that these variants resulted in a decrease in the expression level of PADI6, which may lead to abnormal protein function. Immunofluorescence staining also suggested that these variants affected the expression of PADI6. CONCLUSION Our study expands the spectrum of genetic defects in female early embryo arrest and further supports the causality between PADI6 variants and female infertility.
Collapse
Affiliation(s)
- Juepu Zhou
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Ruolin Mao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Limin Gao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Meng Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Rui Long
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Xiangfei Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Zhou Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China.
| | - Lei Jin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China.
| | - Lixia Zhu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China.
| |
Collapse
|
24
|
Wang X, Bhandari RK. Methylome profile of medaka eggs and sperm. Epigenetics 2024; 19:2417151. [PMID: 39428969 PMCID: PMC11497970 DOI: 10.1080/15592294.2024.2417151] [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: 05/19/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024] Open
Abstract
Eggs and sperm are responsible for the continuation of generations. Following the epigenetic reprogramming of the embryo, core epigenetic information present in the sperm and eggs is transmitted to offspring somatic cells prior to the blastula stage, which specifically influences gene expression in the cells. Differences in the patterns of DNA methylation between the paternal and maternal genomes are critical to regulating allele-specific gene expression in the developing embryo, constituting the basis of genomic imprinting in mammals. While the information on allele-specific epigenetic information has been limited to mammals, it is not clearly understood whether non-mammalian vertebrate gametes possess any sex-specific allelic epigenetic information and whether somatic cells maintain the allele-specific epigenetic information, particularly DNA methylation. To determine the landscape of DNA methylation in paternal and maternal alleles in a non-mammalian vertebrate, we profiled the methylome of egg in medaka fish and compared it with our previously published medaka sperm methylome. We identified a set of gamete-specific differentially methylated regions (DMRs) in the genome- medaka eggs maintained a significantly lower global methylation profile than the sperm. Based on our sequencing depth and data, 10 DMRs were hypermethylated, and 237 DMRs were hypomethylated in the eggs compared to the sperm methylome. Somatic cells in blastula maintained some of those parental gamete-specific DNA methylation profiles. Those DMRs are associated with 70 genes, suggesting that they may have imprinted-like functions and warrant further investigation.
Collapse
Affiliation(s)
- Xuegeng Wang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, College of Life Sciences, South China Normal University, Guangzhou, P. R. China
| | - Ramji K. Bhandari
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA
| |
Collapse
|
25
|
Łuszczki E, Wyszyńska J, Dymek A, Drożdż D, González-Ramos L, Hartgring I, García-Carbonell N, Mazur A, Erdine S, Parnarauskienė J, Alvarez-Pitti J. The Effect of Maternal Diet and Lifestyle on the Risk of Childhood Obesity. Metabolites 2024; 14:655. [PMID: 39728436 DOI: 10.3390/metabo14120655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Childhood obesity is a global health problem that affects at least 41 million children under the age of five. Increased BMI in children is associated with serious long-term health consequences, such as type 2 diabetes, cardiovascular disease, and psychological problems, including depression and low self-esteem. Although the etiology of obesity is complex, research suggests that the diet and lifestyle of pregnant women play a key role in shaping metabolic and epigenetic changes that can increase the risk of obesity in their children. Excessive gestational weight gain, unhealthy dietary patterns (including the Western diet), and pregnancy complications (such as gestational diabetes) are some of the modifiable factors that contribute to childhood obesity. The purpose of this narrative review is to summarize the most important and recent information on the impact of the diet and lifestyle of pregnant women on the risk of childhood obesity. Methods: This article is a narrative review that aims to summarize the available literature on the impact of pregnant women's diet and lifestyle on the risk of obesity in their offspring, with a focus on metabolic and epigenetic mechanisms. Results/Conclusions: Current evidence suggests that a pregnant woman's lifestyle and diet can significantly contribute to lowering the risk of obesity in their offspring. However, further high-quality research is needed to understand better the metabolic and epigenetic relationships concerning maternal factors that predispose offspring to obesity.
Collapse
Affiliation(s)
- Edyta Łuszczki
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Justyna Wyszyńska
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Agnieszka Dymek
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Pediatric Institute, Jagiellonian University Medical College, 31-007 Krakow, Poland
| | - Laura González-Ramos
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Isa Hartgring
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Nuria García-Carbonell
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
| | - Artur Mazur
- Institute of Medical Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Serap Erdine
- Cerrahpasa Faculty of Medicine, Department of Cardiology, Istanbul University-Cerrahpasa, 34320 Istanbul, Turkey
| | - Justė Parnarauskienė
- Pediatric Department, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Julio Alvarez-Pitti
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
26
|
Ilmuratova S, Lokshin V, Prodeus A, Manzhuova L, Nurgaliyeva Z, Kussainova F, Bazarbaeva A, Nekhorosheva V, Abshekenova A. Immune profiling of ART-conceived children in Kazakhstan: a case-control study. Front Pediatr 2024; 12:1447956. [PMID: 39649404 PMCID: PMC11620871 DOI: 10.3389/fped.2024.1447956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/30/2024] [Indexed: 12/10/2024] Open
Abstract
Objective The increasing use of assisted reproductive technologies (ART) has led to a growing interest in the health outcomes of offspring. However, the impact of ART on the immune system of children remains poorly understood. While only two publications were found, their findings contradict each other and did not consider other risk factors in their analysis except for ART use. Therefore, this study aimed to examine the potential impact of ART on the immune system of offspring. Methods A case-control study was conducted in Kazakhstan to investigate the immune system of ART-conceived children compared to those conceived naturally (NC). The study included participants who met certain criteria, such as having undergone a successful ART program resulting in the birth of either a single or multiple pregnancies. Patients who used donor oocytes/sperm, intrauterine insemination, or surrogacy were excluded. Anamnesis data were collected from children in both groups, and laboratory measurements were performed and analyzed using IBM SPSS Statistic 26. Results A total of 120 children conceived by ART and 132 NC children under the age of five were included in our study. We observed that compared with NC group, ART children had lower IgA and IgG levels (p < 0.001), absolute lymphocytosis, high levels of active T-lymphocytes (p = 0.001), and pathological T-helper levels (p = 0.004). Therefore, the clinical presentation of respiratory diseases was lower in ART group. Children born after frozen embryo transfers showed significantly higher levels of T-cytotoxic and active T-lymphocytes compared to children born after fresh embryo transfers (p = 0.007 and p = 0.020, respectively). We utilized ordinal logistic regression to control for confounding variables such as multiple pregnancy, cesarean section, premature birth, and breastfeeding. Despite this, the significant impact of ART on immunogram parameters persisted, indicating the independent and influential nature of ART or other unaccounted factors.
Collapse
Affiliation(s)
- Sevara Ilmuratova
- Department of Science, Kazakhstan Medical University “KSPH”, Almaty, Kazakhstan
| | - Vyacheslav Lokshin
- Department of Assisted Reproductive Technologies, International Clinical Centre of Reproduction “PERSONA”, Almaty, Kazakhstan
| | - Andrey Prodeus
- Science and Education Department, International Academy of Reproductology, Almaty, Kazakhstan
| | - Lyazzat Manzhuova
- Science and Education Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
| | - Zhanar Nurgaliyeva
- Science and Education Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Department of Outpatient Pediatrics, School of Pediatrics, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Farida Kussainova
- Department of Gynecology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Aygul Bazarbaeva
- Science and Education Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
| | - Valeriya Nekhorosheva
- Department of Assisted Reproductive Technologies, Institute of Reproductive Medicine, Almaty, Kazakhstan
| | - Aygerim Abshekenova
- Department of Assisted Reproductive Technologies, International Clinical Centre of Reproduction “PERSONA”, Almaty, Kazakhstan
| |
Collapse
|
27
|
Mori M, Chaudhari BP, Ream MA, Kemper AR. Promises and challenges of genomic newborn screening (NBS) - lessons from public health NBS programs. Pediatr Res 2024:10.1038/s41390-024-03689-0. [PMID: 39516573 DOI: 10.1038/s41390-024-03689-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/03/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Newborn screening (NBS) in the United States began in the 1960s to detect inborn errors of metabolism that benefited from presymptomatic treatment compared with treatment after the development of symptoms and diagnosis. Over time, it expanded to include endocrinological disorders, hematological disorders, immunodeficiencies, and other treatable diseases such as lysosomal storage diseases (LSD), cystic fibrosis, X-linked adrenoleukodystrophy, and spinal muscular dystrophy. This expansion has been driven by new technologies (e.g., tandem mass spectrometry) and novel treatments (e.g., enzyme replacement therapy and stem cell transplant for LSDs). Advances in next-generation gene sequencing (NGS) enable rapid identification of many additional conditions that might benefit from early presymptomatic intervention. We review the NGS technologies that evolved as diagnostic testing and suggest issues to be resolved before their potential application to screening the asymptomatic population. We illustrate the importance of selecting diseases to be screened and propose recommendations to follow when variants of uncertain significance are found. We address ethical issues around achieving equity in the sensitivity of genomic NBS, access to follow-up and management, especially for people from diverse backgrounds, and other considerations. Finally, we discuss the potential benefits and harms of genomic NBS to the overall health of children with monogenic diseases. IMPACT: Genomic newborn screening programs are ongoing worldwide. Public discussion is needed as to whether genomic newborn screening should be offered as a public health program and, if so, what conditions should be screened for. Providers should understand that the sensitivity of genomic newborn screening is especially low for newborns from non-European populations. Methylation, large structural variants and repeat expansion variants are not amenable to next-generation sequencing-based genomic newborn screening. The article serves as a comprehensive guide to understanding issues that need to be solved before genomic newborn screening is implemented as a public health program.
Collapse
Affiliation(s)
- Mari Mori
- The Ohio State University College of Medicine Department of Pediatrics, Columbus, OH, USA.
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
| | - Bimal P Chaudhari
- The Ohio State University College of Medicine Department of Pediatrics, Columbus, OH, USA
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Division of Neonatology, Nationwide Children's Hospital, Columbus, OH, USA
- The Steve and Cindy Rasmussen Institute of Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Margie A Ream
- The Ohio State University College of Medicine Department of Pediatrics, Columbus, OH, USA
- Division of Division of Child Neurology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Alex R Kemper
- The Ohio State University College of Medicine Department of Pediatrics, Columbus, OH, USA
- Division of Primary Care Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
28
|
Cervantes-Salazar JL, Pérez-Hernández N, Calderón-Colmenero J, Rodríguez-Pérez JM, González-Pacheco MG, Villamil-Castañeda C, Rosas-Tlaque AA, Ortega-Zhindón DB. Genetic Insights into Congenital Cardiac Septal Defects-A Narrative Review. BIOLOGY 2024; 13:911. [PMID: 39596866 PMCID: PMC11592229 DOI: 10.3390/biology13110911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Congenital heart diseases (CHDs) are a group of complex diseases characterized by structural and functional malformations during development in the human heart; they represent an important problem for public health worldwide. Within these malformations, septal defects such as ventricular (VSD) and atrial septal defects (ASD) are the most common forms of CHDs. Studies have reported that CHDs are the result of genetic and environmental factors. Here, we review and summarize the role of genetics involved in cardiogenesis and congenital cardiac septal defects. Moreover, treatment regarding these congenital cardiac septal defects is also addressed.
Collapse
Affiliation(s)
- Jorge L. Cervantes-Salazar
- Department of Pediatric Cardiac Surgery and Congenital Heart Disease, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (J.L.C.-S.); (A.A.R.-T.)
| | - Nonanzit Pérez-Hernández
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (N.P.-H.); (J.M.R.-P.); (C.V.-C.)
| | - Juan Calderón-Colmenero
- Department of Pediatric Cardiology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - José Manuel Rodríguez-Pérez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (N.P.-H.); (J.M.R.-P.); (C.V.-C.)
| | | | - Clara Villamil-Castañeda
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (N.P.-H.); (J.M.R.-P.); (C.V.-C.)
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Angel A. Rosas-Tlaque
- Department of Pediatric Cardiac Surgery and Congenital Heart Disease, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (J.L.C.-S.); (A.A.R.-T.)
- Dirección General de Calidad y Educación en Salud, Secretaría de Salud, Mexico City 06600, Mexico
| | - Diego B. Ortega-Zhindón
- Department of Pediatric Cardiac Surgery and Congenital Heart Disease, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (J.L.C.-S.); (A.A.R.-T.)
- Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
29
|
Urakawa T, Soejima H, Yamoto K, Hara-Isono K, Nakamura A, Kawashima S, Narusawa H, Kosaki R, Nishimura Y, Yamazawa K, Hattori T, Muramatsu Y, Inoue T, Matsubara K, Fukami M, Saitoh S, Ogata T, Kagami M. Comprehensive molecular and clinical findings in 29 patients with multi-locus imprinting disturbance. Clin Epigenetics 2024; 16:138. [PMID: 39369220 PMCID: PMC11452994 DOI: 10.1186/s13148-024-01744-5] [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: 05/13/2024] [Accepted: 09/13/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Multi-locus imprinting disturbance (MLID) with methylation defects in various differentially methylated regions (DMRs) has recently been identified in approximately 150 cases with imprinting disorders (IDs), and deleterious variants have been found in genes related to methylation maintenance of DMRs, such as those encoding proteins constructing the subcortical maternal complex (SCMC), in a small fraction of patients and/or their mothers. However, integrated methylation analysis for DMRs and sequence analysis for MLID-causative genes in MLID cases and their mothers have been performed only in a single study focusing on Beckwith-Wiedemann syndrome (BWS) and Silver-Russell syndrome (SRS) phenotypes. RESULTS Of 783 patients with various IDs we have identified to date, we examined a total of 386 patients with confirmed epimutation and 71 patients with epimutation or uniparental disomy. Consequently, we identified MLID in 29 patients with epimutation confirmed by methylation analysis for multiple ID-associated DMRs using pyrosequencing and/or methylation-specific multiple ligation-dependent probe amplification. MLID was detected in approximately 12% of patients with BWS phenotype and approximately 5% of patients with SRS phenotype, but not in patients with Kagami-Ogata syndrome, Prader-Willi syndrome, or Angelman syndrome phenotypes. We next conducted array-based methylation analysis for 78 DMRs and whole-exome sequencing in the 29 patients, revealing hypomethylation-dominant aberrant methylation patterns in various DMRs of all the patients, eight probably deleterious variants in genes for SCMC in the mothers of patients, and one homozygous deleterious variant in ZNF445 in one patient. These variants did not show gene-specific methylation disturbance patterns. Clinically, neurodevelopmental delay and/or intellectual developmental disorder (ND/IDD) was observed in about half of the MLID patients, with no association with the identified methylation disturbance patterns and genetic variants. Notably, seven patients with BWS phenotype were conceived by assisted reproductive technology (ART). CONCLUSIONS The frequency of MLID was 7.5% (29/386) in IDs caused by confirmed epimutation. Furthermore, we revealed diverse patterns of hypomethylation-dominant methylation defects, nine deleterious variants, ND/IDD complications in about half of the MLID patients, and a high frequency of MLID in ART-conceived patients.
Collapse
Affiliation(s)
- Tatsuki Urakawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
- Department of Pediatrics, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8102, Japan
| | - Hidenobu Soejima
- Division of Molecular Genetics and Epigenetics, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-0937, Japan
| | - Kaori Yamoto
- Department of Biochemistry, Hamamatsu University School of Medicine, 1‑20‑1 Handayama, Higashi‑ku, Hamamatsu, 431‑3192, Japan
| | - Kaori Hara-Isono
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Akie Nakamura
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Sayaka Kawashima
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Hiromune Narusawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Rika Kosaki
- Department of Medical Genetics, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Yutaka Nishimura
- Department of Neonatology, Hiroshima City Hiroshima Citizens Hospital, 7-33 Motomachi, Naka-Ku, Hiroshima, 730-8518, Japan
| | - Kazuki Yamazawa
- Medical Genetics Center, NHO Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-Ku, Tokyo, 152-8902, Japan
| | - Tetsuo Hattori
- Department of Pediatrics, Anjo Kosei Hospital, 28 Higashihirokute, Anjo, 446-8602, Japan
| | - Yukako Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Shouwa‑ku, Nagoya, 466‑8560, Japan
| | - Takanobu Inoue
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta, Midori-Ku, Chiba, 266-0007, Japan
| | - Keiko Matsubara
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
- Division of Diversity Research, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Tsutomu Ogata
- Department of Biochemistry, Hamamatsu University School of Medicine, 1‑20‑1 Handayama, Higashi‑ku, Hamamatsu, 431‑3192, Japan
- Department of Pediatrics, Hamamatsu Medical Center, 328 Tomizuka-Cho, Chuo-Ku, Hamamatsu, 432-8580, Japan
| | - Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan.
| |
Collapse
|
30
|
Esteller M, Dawson MA, Kadoch C, Rassool FV, Jones PA, Baylin SB. The Epigenetic Hallmarks of Cancer. Cancer Discov 2024; 14:1783-1809. [PMID: 39363741 DOI: 10.1158/2159-8290.cd-24-0296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/08/2024] [Accepted: 06/24/2024] [Indexed: 10/05/2024]
Abstract
Cancer is a complex disease in which several molecular and cellular pathways converge to foster the tumoral phenotype. Notably, in the latest iteration of the cancer hallmarks, "nonmutational epigenetic reprogramming" was newly added. However, epigenetics, much like genetics, is a broad scientific area that deserves further attention due to its multiple roles in cancer initiation, progression, and adaptive nature. Herein, we present a detailed examination of the epigenetic hallmarks affected in human cancer, elucidating the pathways and genes involved, and dissecting the disrupted landscapes for DNA methylation, histone modifications, and chromatin architecture that define the disease. Significance: Cancer is a disease characterized by constant evolution, spanning from its initial premalignant stages to the advanced invasive and disseminated stages. It is a pathology that is able to adapt and survive amidst hostile cellular microenvironments and diverse treatments implemented by medical professionals. The more fixed setup of the genetic structure cannot fully provide transformed cells with the tools to survive but the rapid and plastic nature of epigenetic changes is ready for the task. This review summarizes the epigenetic hallmarks that define the ecological success of cancer cells in our bodies.
Collapse
Affiliation(s)
- Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
| | - Mark A Dawson
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, Australia
| | - Cigall Kadoch
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Feyruz V Rassool
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peter A Jones
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan
| | - Stephen B Baylin
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| |
Collapse
|
31
|
Giaccari C, Cecere F, Argenziano L, Pagano A, Riccio A. New insights into oocyte cytoplasmic lattice-associated proteins. Trends Genet 2024; 40:880-890. [PMID: 38955588 DOI: 10.1016/j.tig.2024.06.002] [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: 04/16/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024]
Abstract
Oocyte maturation and preimplantation embryo development are critical to successful pregnancy outcomes and the correct establishment and maintenance of genomic imprinting. Thanks to novel technologies and omics studies in human patients and mouse models, the importance of the proteins associated with the cytoplasmic lattices (CPLs), highly abundant structures found in the cytoplasm of mammalian oocytes and preimplantation embryos, in the maternal to zygotic transition is becoming increasingly evident. This review highlights the recent discoveries on the role of these proteins in protein storage and other oocyte cytoplasmic processes, epigenetic reprogramming, and zygotic genome activation (ZGA). A better comprehension of these events may significantly improve clinical diagnosis and pave the way for targeted interventions aiming to correct or mitigate female fertility issues and genomic imprinting disorders.
Collapse
Affiliation(s)
- Carlo Giaccari
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania 'Luigi Vanvitelli,' Caserta, Italy
| | - Francesco Cecere
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania 'Luigi Vanvitelli,' Caserta, Italy
| | - Lucia Argenziano
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania 'Luigi Vanvitelli,' Caserta, Italy
| | - Angela Pagano
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania 'Luigi Vanvitelli,' Caserta, Italy
| | - Andrea Riccio
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania 'Luigi Vanvitelli,' Caserta, Italy; Institute of Genetics and Biophysics (IGB) 'Adriano Buzzati-Traverso,' Consiglio Nazionale delle Ricerche (CNR), Naples, Italy.
| |
Collapse
|
32
|
Ye M, Reyes Palomares A, Iwarsson E, Oberg AS, Rodriguez-Wallberg KA. Imprinting disorders in children conceived with assisted reproductive technology in Sweden. Fertil Steril 2024; 122:706-714. [PMID: 38825304 DOI: 10.1016/j.fertnstert.2024.05.168] [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: 12/27/2023] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/04/2024]
Abstract
OBJECTIVE To assess whether the use of assisted reproductive technology (ART) therapy for conception is associated with imprinting disorders in children and the impact of parental factors related to infertility. DESIGN A nationwide register-based cohort study. SETTING Swedish national registers and nationwide quality IVF register. PATIENT(S) All liveborn singletons in Sweden (N = 2,084,127) between 1997 and 2017 with follow-up to December 31, 2018. INTERVENTION(S) The use of specific methods implemented in ART. MAIN OUTCOME MEASURE(S) The International Classification of Diseases version 10 was used to identify three distinct imprinting disorder groups: Beckwith-Wiedemann syndrome (BWS), Prader-Willi syndrome (PWS), and Silver-Russell syndrome (SRS), as well as central precocious puberty. The Cox model combined with inverse probability treatment weights was used to estimate the weighted hazard ratio (wHR) with a 95% confidence interval (CI), accounting for multiple confounders. RESULT(S) A total of 1,044 children were diagnosed with the disorders of interest, and 52 of them were conceived using ART therapy. The overall risk of being diagnosed with any of the studied imprinting disorders was elevated in children conceived using ART therapy compared with all other children (HR, 1.84; 95% CI, 1.38-2.45). After adjusting for parental background factors, the association was partially attenuated (wHR, 1.50; 95% CI, 0.97-2.32), but remained in the weighted comparison restricted to children of couples with known infertility (wHR, 1.52; 95% CI, 1.05-2.21). For the specific diagnoses of PWS/SRS, and BWS compared with children of couples with known infertility, children conceived with ART therapy showed a small excess risk, which could not be distinguished from the null (wHR, 1.56; 95% CI, 0.93-2.62 and 1.80; 95% CI, 0.99-3.28, respectively). Further subgroup analysis showed that the combined use of intracytoplasmic sperm injection and cryopreserved embryos was associated with a higher risk of both PWS/SRS (wHR, 4.60; 95% CI, 1.72-12.28) and BWS (wHR, 6.69; 95% CI, 2.09-21.45). The number of central precocious puberty cases in children conceived using ART therapy was too small (N = 3) to make any meaningful inference. CONCLUSION(S) The combined use of intracytoplasmic sperm injection and cryopreserved embryos was associated with small elevated risks of PWS/SRS, and BWS in children, independent of parental factors related to infertility.
Collapse
Affiliation(s)
- Mujin Ye
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Erik Iwarsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna S Oberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Division of Gynecology and Reproduction, Department of Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
33
|
Kaur R, Rishi V. Transcription factors and genome biases in polyploid crops. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 143:301-321. [PMID: 39843138 DOI: 10.1016/bs.apcsb.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Nuclear protein transcription factors (TFs) regulate all biological processes in plants and are necessary for gene regulation. The transcription of genes during plant growth and development and their response to environmental cues are regulated by TF binding to specific promoter regions in the genomic DNA. Polyploid plants with several sets of chromosomes frequently display intricate genomic biases concerning TF expression. One or more subgenomes may dominate in terms of gene expression, leading to subgenome biases or dominance. These biases can influence various aspects of the crop's biology, including its growth, development, and adaptation. Advances in genomics have speed up the improvement of many important agricultural diploid crops, yet comparable endeavours in polyploid crops have been more challenging. This challenge primarily stems from the large size and intricate nature of the complex genome in polyploid crops, along with the need for comprehensive genome assembly data for such crop varieties as bread wheat, cotton and sugarcane. Several studies have evaluated the biased/asymmetric gene expression patterns, including TFs, within the polyploid crop genomes. In many polyploid crops, not all homologues of TF genes contribute equally to the phenotype. Here, we have examined polyploid crop plants for homeolog gene expression, emphasizing TFs. It is observed that the polyploids retain many gene alleles as functional homeologs that define important features involved in stress response, sugar metabolism, and fibre formation. The possible molecular mechanism describing the structural and epigenetic basis of differential subgenomic TF expression in polyploids is discussed.
Collapse
Affiliation(s)
- Raminder Kaur
- National Agri-Food Biotechnology Institute, Knowledge City, Mohali, Punjab, India
| | - Vikas Rishi
- National Agri-Food Biotechnology Institute, Knowledge City, Mohali, Punjab, India.
| |
Collapse
|
34
|
Nomani H, Wu S, Saif A, Hwang F, Metzger J, Navetta-Modrov B, Gorevic PD, Aksentijevich I, Yao Q. Comprehensive clinical phenotype, genotype and therapy in Yao syndrome. Front Immunol 2024; 15:1458118. [PMID: 39372397 PMCID: PMC11449693 DOI: 10.3389/fimmu.2024.1458118] [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: 07/01/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
Objective Yao syndrome (YAOS) is formerly called nucleotide-binding oligomerization domain containing 2 (NOD2)-associated autoinflammatory disease.We report a large cohort of YAOS. Methods We conducted a retrospective analysis of a cohort of adult patients with systemic autoinflammatory diseases (SAIDs). All patients underwent testing for a periodic fever syndrome gene panel. Results A total of 194 patients carried NOD2 variants, 152 patients were diagnosed with YAOS, and 42 had mixed autoinflammatory diseases with combined variants in NOD2 and other SAID-associated genes. Demographic, clinical and molecular data were summaried. In sub-group analysis of the 194 patients, individual patients were often identified to carry two or more variants that usually included IVS8 + 158/R702W, IVS8 + 158/L1007fs, IVS8 + 158/V955I, IVS8 + 158/other, or NOD2/variants in other SAID genes. Ninety-nine patients carried single variants. Taken together, these variants contribute to the disease in combination or individually. Conclusion This largest cohort has provided comprehensive clinical and genotyping data in YAOS. Variants in the NOD2 gene can give rise to a spectrum from inflammatory bowel disease to autoinflammatory disease.This report further raises awareness of the underdiagnosed disease in the medical community.
Collapse
Affiliation(s)
- Hafsa Nomani
- Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, United States
| | - Song Wu
- Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, United States
| | - Ashmia Saif
- Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, United States
| | - Frank Hwang
- Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, United States
| | - Jane Metzger
- Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, United States
| | - Brianne Navetta-Modrov
- Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, United States
| | - Peter D. Gorevic
- Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, United States
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Qingping Yao
- Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, United States
| |
Collapse
|
35
|
Bækgaard CH, Lester EB, Møller-Larsen S, Lauridsen MF, Larsen MJ. NanoImprint: A DNA methylation tool for clinical interpretation and diagnosis of common imprinting disorders using nanopore long-read sequencing. Ann Hum Genet 2024; 88:392-398. [PMID: 38690755 DOI: 10.1111/ahg.12556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/15/2024] [Accepted: 02/23/2024] [Indexed: 05/03/2024]
Abstract
INTRODUCTION Long-read whole genome sequencing like Oxford Nanopore Technology, is increasingly being introduced in clinical settings. With its ability to simultaneously call sequence variation and DNA modifications including 5-methylcytosine, nanopore is a promising technology to improve diagnostics of imprinting disorders. METHODS Currently, no tools to analyze DNA methylation patterns at known clinically relevant imprinted regions are available. Here we present NanoImprint, which generates an easily interpretable report, based on long-read nanopore sequencing, to use for identifying clinical relevant abnormalities in methylation levels at 14 imprinted regions and diagnosis of common imprinting disorders. RESULTS AND CONCLUSION NanoImprint outputs a summarizing table and visualization plots displays methylation frequency (%) and chromosomal positions for all regions, with phased data color-coded for the two alleles. We demonstrate the utility of NanoImprint using three imprinting disorder samples from patients with Beckwith-Wiedemann syndrome (BWS), Angelman syndrome (AS) and Prader-Willi syndrome (PWS). NanoImprint script is available from https://github.com/carolinehey/NanoImprint.
Collapse
Affiliation(s)
| | - Emilie Boye Lester
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Steffen Møller-Larsen
- Clinical Genome Center, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | - Martin Jakob Larsen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
36
|
Li N, Liu HY, Liu SM. Deciphering DNA Methylation in Gestational Diabetes Mellitus: Epigenetic Regulation and Potential Clinical Applications. Int J Mol Sci 2024; 25:9361. [PMID: 39273309 PMCID: PMC11394902 DOI: 10.3390/ijms25179361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Gestational diabetes mellitus (GDM) represents a prevalent complication during pregnancy, exerting both short-term and long-term impacts on maternal and offspring health. This review offers a comprehensive outline of DNA methylation modifications observed in various maternal and offspring tissues affected by GDM, emphasizing the intricate interplay between DNA methylation dynamics, gene expression, and the pathogenesis of GDM. Furthermore, it explores the influence of environmental pollutants, maternal nutritional supplementation, and prenatal gut microbiota on GDM development through alterations in DNA methylation profiles. Additionally, this review summarizes recent advancements in DNA methylation-based diagnostics and predictive models in early GDM detection and risk assessment for subsequent type 2 diabetes. These insights contribute significantly to our understanding of the epigenetic mechanisms underlying GDM development, thereby enhancing maternal and fetal health outcomes and advocating further efforts in this field.
Collapse
Affiliation(s)
- Nan Li
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Huan-Yu Liu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, 169 Donghu Road, Wuhan 430071, China
| | - Song-Mei Liu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, 169 Donghu Road, Wuhan 430071, China
| |
Collapse
|
37
|
Joshi DC, Sharma A, Prasad S, Singh K, Kumar M, Sherawat K, Tuli HS, Gupta M. Novel therapeutic agents in clinical trials: emerging approaches in cancer therapy. Discov Oncol 2024; 15:342. [PMID: 39127974 PMCID: PMC11317456 DOI: 10.1007/s12672-024-01195-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Novel therapeutic agents in clinical trials offer a paradigm shift in the approach to battling this prevalent and destructive disease, and the area of cancer therapy is on the precipice of a trans formative revolution. Despite the importance of tried-and-true cancer treatments like surgery, radiation, and chemotherapy, the disease continues to evolve and adapt, making new, more potent methods necessary. The field of cancer therapy is currently witnessing the emergence of a wide range of innovative approaches. Immunotherapy, including checkpoint inhibitors, CAR-T cell treatment, and cancer vaccines, utilizes the host's immune system to selectively target and eradicate malignant cells while minimizing harm to normal tissue. The development of targeted medicines like kinase inhibitors and monoclonal antibodies has allowed for more targeted and less harmful approaches to treating cancer. With the help of genomics and molecular profiling, "precision medicine" customizes therapies to each patient's unique genetic makeup to maximize therapeutic efficacy while minimizing unwanted side effects. Epigenetic therapies, metabolic interventions, radio-pharmaceuticals, and an increasing emphasis on combination therapy with synergistic effects further broaden the therapeutic landscape. Multiple-stage clinical trials are essential for determining the safety and efficacy of these novel drugs, allowing patients to gain access to novel treatments while also furthering scientific understanding. The future of cancer therapy is rife with promise, as the integration of artificial intelligence and big data has the potential to revolutionize early detection and prevention. Collaboration among researchers, and healthcare providers, and the active involvement of patients remain the bedrock of the ongoing battle against cancer. In conclusion, the dynamic and evolving landscape of cancer therapy provides hope for improved treatment outcomes, emphasizing a patient-centered, data-driven, and ethically grounded approach as we collectively strive towards a cancer-free world.
Collapse
Affiliation(s)
- Deepak Chandra Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Dist., Ajmer, Rajasthan, India.
| | - Anurag Sharma
- Invertis Institute of Pharmacy, Invertis University Bareilly Uttar Pradesh, Bareilly, India
| | - Sonima Prasad
- Chandigarh University, Ludhiana-Chandigarh State Highway, Gharuan, Mohali, Punjab, 140413, India
| | - Karishma Singh
- Institute of Pharmaceutical Sciences, Faculty of Engineering and Technology, University of Lucknow, Lucknow, India
| | - Mayank Kumar
- Himalayan Institute of Pharmacy, Road, Near Suketi Fossil Park, Kala Amb, Hamidpur, Himachal Pradesh, India
| | - Kajal Sherawat
- Meerut Institute of Technology, Meerut, Uttar Pradesh, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences & Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, India
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India.
| |
Collapse
|
38
|
Holthöfer L, Diederich S, Haug V, Lehmann L, Hewel C, Paul NW, Schweiger S, Gerber S, Linke M. A case of an Angelman-syndrome caused by an intragenic duplication of UBE3A uncovered by adaptive nanopore sequencing. Clin Epigenetics 2024; 16:101. [PMID: 39095842 PMCID: PMC11297752 DOI: 10.1186/s13148-024-01711-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Adaptive nanopore sequencing as a diagnostic method for imprinting disorders and episignature analysis revealed an intragenic duplication of Exon 6 and 7 in UBE3A (NM_000462.5) in a patient with relatively mild Angelman-like syndrome. In an all-in-one nanopore sequencing analysis DNA hypomethylation of the SNURF:TSS-DMR, known contributing deletions on the maternal allele and point mutations in UBE3A could be ruled out as disease drivers. In contrast, breakpoints and orientation of the tandem duplication could clearly be defined. Segregation analysis in the family showed that the duplication derived de novo in the maternal grandfather. Our study shows the benefits of an all-in-one nanopore sequencing approach for the diagnostics of Angelman syndrome and other imprinting disorders.
Collapse
Affiliation(s)
- Laura Holthöfer
- Institute for Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Diederich
- Institute for Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Verena Haug
- Neuropediatrics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lioba Lehmann
- Institute for Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Charlotte Hewel
- Institute for Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Norbert W Paul
- Institute for History, Philosophy, and Ethics of Medicine, Johannes Gutenberg-University Medical Center Mainz, Mainz, Germany
| | - Susann Schweiger
- Institute for Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Susanne Gerber
- Institute for Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Linke
- Institute for Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
39
|
Mackay DJG, Gazdagh G, Monk D, Brioude F, Giabicani E, Krzyzewska IM, Kalish JM, Maas SM, Kagami M, Beygo J, Kahre T, Tenorio-Castano J, Ambrozaitytė L, Burnytė B, Cerrato F, Davies JH, Ferrero GB, Fjodorova O, Manero-Azua A, Pereda A, Russo S, Tannorella P, Temple KI, Õunap K, Riccio A, de Nanclares GP, Maher ER, Lapunzina P, Netchine I, Eggermann T, Bliek J, Tümer Z. Multi-locus imprinting disturbance (MLID): interim joint statement for clinical and molecular diagnosis. Clin Epigenetics 2024; 16:99. [PMID: 39090763 PMCID: PMC11295890 DOI: 10.1186/s13148-024-01713-y] [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: 04/25/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Imprinting disorders are rare diseases resulting from altered expression of imprinted genes, which exhibit parent-of-origin-specific expression patterns regulated through differential DNA methylation. A subgroup of patients with imprinting disorders have DNA methylation changes at multiple imprinted loci, a condition referred to as multi-locus imprinting disturbance (MLID). MLID is recognised in most but not all imprinting disorders and is also found in individuals with atypical clinical features; the presence of MLID often alters the management or prognosis of the affected person. Some cases of MLID are caused by trans-acting genetic variants, frequently not in the patients but their mothers, which have counselling implications. There is currently no consensus on the definition of MLID, clinical indications prompting testing, molecular procedures and methods for epigenetic and genetic diagnosis, recommendations for laboratory reporting, considerations for counselling, and implications for prognosis and management. The purpose of this study is thus to cover this unmet need. METHODS A comprehensive literature search was conducted resulting in identification of more than 100 articles which formed the basis of discussions by two working groups focusing on clinical diagnosis (n = 12 members) and molecular testing (n = 19 members). Following eight months of preparations and regular online discussions, the experts from 11 countries compiled the preliminary documentation and determined the questions to be addressed during a face-to-face meeting which was held with the attendance of the experts together with four representatives of patient advocacy organisations. RESULTS In light of available evidence and expert consensus, we formulated 16 propositions and 8 recommendations as interim guidance for the clinical and molecular diagnosis of MLID. CONCLUSIONS MLID is a molecular designation, and for patients with MLID and atypical phenotypes, we propose the alternative term multi-locus imprinting syndrome. Due to the intrinsic variability of MLID, the guidelines underscore the importance of involving experts from various fields to ensure a confident approach to diagnosis, counselling, and care. The authors advocate for global, collaborative efforts in both basic and translational research to tackle numerous crucial questions that currently lack answers, and suggest reconvening within the next 3-5 years to evaluate the research advancements and update this guidance as needed.
Collapse
Affiliation(s)
| | - Gabriella Gazdagh
- Faculty of Medicine, University of Southampton, Southampton, UK
- Wessex Clinical Genetics Service, Princess Anne Hospital, University Hospital Southampton NHS Trust, Southampton, UK
| | - David Monk
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Frederic Brioude
- Centre de Recherche Saint Antoine, Endocrinologie Moléculaire et Pathologies d'empreinte, INSERMSorbonne Université, Hôpital Armand TrousseauAPHP, 75012, Paris, France
| | - Eloise Giabicani
- Centre de Recherche Saint Antoine, Endocrinologie Moléculaire et Pathologies d'empreinte, INSERMSorbonne Université, Hôpital Armand TrousseauAPHP, 75012, Paris, France
| | - Izabela M Krzyzewska
- Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jennifer M Kalish
- Division of Human Genetics and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Departments of Pediatrics and Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Saskia M Maas
- Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Jasmin Beygo
- Institut Für Humangenetik, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Tiina Kahre
- Department of Laboratory Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Jair Tenorio-Castano
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- Institute of Medical and Molecular Genetics, INGEMM-Idipaz, Madrid, Spain
| | - Laima Ambrozaitytė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Birutė Burnytė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Flavia Cerrato
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università Degli Studi Della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Justin H Davies
- Faculty of Medicine, University of Southampton, Southampton, UK
- Regional Centre for Paediatric Endocrinology, Faculty of Medicine, Southampton Children's Hospital, University of Southampton, Southampton, UK
| | - Giovanni Battista Ferrero
- Department of Clinical and Biological Science, School of Medicine, Centre for Hemoglobinopathies, AOU San Luigi Gonzaga, University of Turin, Turin, Italy
| | - Olga Fjodorova
- Department of Laboratory Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Africa Manero-Azua
- Rare Diseases Research Group, Molecular (Epi)Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Araba, Spain
| | - Arrate Pereda
- Rare Diseases Research Group, Molecular (Epi)Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Araba, Spain
| | - Silvia Russo
- IRCCS Research Laboratory of Medical Cytogenetics and Molecular Genetics, Istituto Auxologico Italiano, Milan, Italy
| | - Pierpaola Tannorella
- IRCCS Research Laboratory of Medical Cytogenetics and Molecular Genetics, Istituto Auxologico Italiano, Milan, Italy
| | - Karen I Temple
- Faculty of Medicine, University of Southampton, Southampton, UK
- Wessex Clinical Genetics Service, Princess Anne Hospital, University Hospital Southampton NHS Trust, Southampton, UK
| | - Katrin Õunap
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Department of Clinical Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Andrea Riccio
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università Degli Studi Della Campania "Luigi Vanvitelli", Caserta, Italy
- Institute of Genetics and Biophysics (IGB),"Adriano Buzzati-Traverso", Consiglio Nazionale Delle Ricerche (CNR), Naples, Italy
| | - Guiomar Perez de Nanclares
- Rare Diseases Research Group, Molecular (Epi)Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Araba, Spain
| | - Eamonn R Maher
- Aston Medical School, Aston University, Birmingham, UK
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Pablo Lapunzina
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- Institute of Medical and Molecular Genetics, INGEMM-Idipaz, Madrid, Spain
| | - Irène Netchine
- Centre de Recherche Saint Antoine, Endocrinologie Moléculaire et Pathologies d'empreinte, INSERMSorbonne Université, Hôpital Armand TrousseauAPHP, 75012, Paris, France
| | - Thomas Eggermann
- Institute for Human Genetics and Genome Medicine. Faculty of Medicine, RWTH University Aachen, Aachen, Germany
| | - Jet Bliek
- Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Zeynep Tümer
- Department of Clinical Genetics, Kennedy Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Stefansson OA, Sigurpalsdottir BD, Rognvaldsson S, Halldorsson GH, Juliusson K, Sveinbjornsson G, Gunnarsson B, Beyter D, Jonsson H, Gudjonsson SA, Olafsdottir TA, Saevarsdottir S, Magnusson MK, Lund SH, Tragante V, Oddsson A, Hardarson MT, Eggertsson HP, Gudmundsson RL, Sverrisson S, Frigge ML, Zink F, Holm H, Stefansson H, Rafnar T, Jonsdottir I, Sulem P, Helgason A, Gudbjartsson DF, Halldorsson BV, Thorsteinsdottir U, Stefansson K. The correlation between CpG methylation and gene expression is driven by sequence variants. Nat Genet 2024; 56:1624-1631. [PMID: 39048797 PMCID: PMC11319203 DOI: 10.1038/s41588-024-01851-2] [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: 04/13/2022] [Accepted: 06/27/2024] [Indexed: 07/27/2024]
Abstract
Gene promoter and enhancer sequences are bound by transcription factors and are depleted of methylated CpG sites (cytosines preceding guanines in DNA). The absence of methylated CpGs in these sequences typically correlates with increased gene expression, indicating a regulatory role for methylation. We used nanopore sequencing to determine haplotype-specific methylation rates of 15.3 million CpG units in 7,179 whole-blood genomes. We identified 189,178 methylation depleted sequences where three or more proximal CpGs were unmethylated on at least one haplotype. A total of 77,789 methylation depleted sequences (~41%) associated with 80,503 cis-acting sequence variants, which we termed allele-specific methylation quantitative trait loci (ASM-QTLs). RNA sequencing of 896 samples from the same blood draws used to perform nanopore sequencing showed that the ASM-QTL, that is, DNA sequence variability, drives most of the correlation found between gene expression and CpG methylation. ASM-QTLs were enriched 40.2-fold (95% confidence interval 32.2, 49.9) among sequence variants associating with hematological traits, demonstrating that ASM-QTLs are important functional units in the noncoding genome.
Collapse
Affiliation(s)
| | - Brynja Dogg Sigurpalsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | | | - Gisli Hreinn Halldorsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | | | | | | | - Thorunn Asta Olafsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Saedis Saevarsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Magnus Karl Magnusson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Sigrun Helga Lund
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | - Marteinn Thor Hardarson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | | | | | | | | | | | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | | | | | - Ingileif Jonsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Agnar Helgason
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Department of Anthropology, University of Iceland, Reykjavik, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Bjarni V Halldorsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
41
|
Farhadova S, Ghousein A, Charon F, Surcis C, Gomez-Velazques M, Roidor C, Di Michele F, Borensztein M, De Sario A, Esnault C, Noordermeer D, Moindrot B, Feil R. The long non-coding RNA Meg3 mediates imprinted gene expression during stem cell differentiation. Nucleic Acids Res 2024; 52:6183-6200. [PMID: 38613389 PMCID: PMC11194098 DOI: 10.1093/nar/gkae247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/02/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
The imprinted Dlk1-Dio3 domain comprises the developmental genes Dlk1 and Rtl1, which are silenced on the maternal chromosome in different cell types. On this parental chromosome, the domain's imprinting control region activates a polycistron that produces the lncRNA Meg3 and many miRNAs (Mirg) and C/D-box snoRNAs (Rian). Although Meg3 lncRNA is nuclear and associates with the maternal chromosome, it is unknown whether it controls gene repression in cis. We created mouse embryonic stem cells (mESCs) that carry an ectopic poly(A) signal, reducing RNA levels along the polycistron, and generated Rian-/- mESCs as well. Upon ESC differentiation, we found that Meg3 lncRNA (but not Rian) is required for Dlk1 repression on the maternal chromosome. Biallelic Meg3 expression acquired through CRISPR-mediated demethylation of the paternal Meg3 promoter led to biallelic Dlk1 repression, and to loss of Rtl1 expression. lncRNA expression also correlated with DNA hypomethylation and CTCF binding at the 5'-side of Meg3. Using Capture Hi-C, we found that this creates a Topologically Associating Domain (TAD) organization that brings Meg3 close to Dlk1 on the maternal chromosome. The requirement of Meg3 for gene repression and TAD structure may explain how aberrant MEG3 expression at the human DLK1-DIO3 locus associates with imprinting disorders.
Collapse
Affiliation(s)
- Sabina Farhadova
- Institute of Molecular Genetics of Montpellier (IGMM), Centre National de Recherche Scientifique (CNRS), 34090 Montpellier, France
- University of Montpellier, 34090 Montpellier, France
- Genetic Resources Research Institute, Azerbaijan National Academy of Sciences (ANAS), AZ1106 Baku, Azerbaijan
| | - Amani Ghousein
- Institute of Molecular Genetics of Montpellier (IGMM), Centre National de Recherche Scientifique (CNRS), 34090 Montpellier, France
- University of Montpellier, 34090 Montpellier, France
| | - François Charon
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190 Gif-sur-Yvette, France
| | - Caroline Surcis
- Institute of Molecular Genetics of Montpellier (IGMM), Centre National de Recherche Scientifique (CNRS), 34090 Montpellier, France
| | - Melisa Gomez-Velazques
- Institute of Molecular Genetics of Montpellier (IGMM), Centre National de Recherche Scientifique (CNRS), 34090 Montpellier, France
- University of Montpellier, 34090 Montpellier, France
| | - Clara Roidor
- Institute of Molecular Genetics of Montpellier (IGMM), Centre National de Recherche Scientifique (CNRS), 34090 Montpellier, France
- University of Montpellier, 34090 Montpellier, France
| | - Flavio Di Michele
- Institute of Molecular Genetics of Montpellier (IGMM), Centre National de Recherche Scientifique (CNRS), 34090 Montpellier, France
- University of Montpellier, 34090 Montpellier, France
| | - Maud Borensztein
- Institute of Molecular Genetics of Montpellier (IGMM), Centre National de Recherche Scientifique (CNRS), 34090 Montpellier, France
- University of Montpellier, 34090 Montpellier, France
| | - Albertina De Sario
- University of Montpellier, 34090 Montpellier, France
- PhyMedExp, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS, 34295 Montpellier, France
| | - Cyril Esnault
- Institute of Molecular Genetics of Montpellier (IGMM), Centre National de Recherche Scientifique (CNRS), 34090 Montpellier, France
- University of Montpellier, 34090 Montpellier, France
| | - Daan Noordermeer
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190 Gif-sur-Yvette, France
| | - Benoit Moindrot
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91190 Gif-sur-Yvette, France
| | - Robert Feil
- Institute of Molecular Genetics of Montpellier (IGMM), Centre National de Recherche Scientifique (CNRS), 34090 Montpellier, France
- University of Montpellier, 34090 Montpellier, France
| |
Collapse
|
42
|
Matsuzaki H, Kimura M, Morihashi M, Tanimoto K. Imprinted DNA methylation of the H19 ICR is established and maintained in vivo in the absence of Kaiso. Epigenetics Chromatin 2024; 17:20. [PMID: 38840164 PMCID: PMC11151560 DOI: 10.1186/s13072-024-00544-8] [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: 02/03/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Paternal allele-specific DNA methylation of the imprinting control region (H19 ICR) controls genomic imprinting at the Igf2/H19 locus. We previously demonstrated that the mouse H19 ICR transgene acquires imprinted DNA methylation in preimplantation mouse embryos. This activity is also present in the endogenous H19 ICR and protects it from genome-wide reprogramming after fertilization. We also identified a 118-bp sequence within the H19 ICR that is responsible for post-fertilization imprinted methylation. Two mutations, one in the five RCTG motifs and the other a 36-bp deletion both in the 118-bp segment, caused complete and partial loss, respectively, of methylation following paternal transmission in each transgenic mouse. Interestingly, these mutations overlap with the binding site for the transcription factor Kaiso, which is reportedly involved in maintaining paternal methylation at the human H19 ICR (IC1) in cultured cells. In this study, we investigated if Kaiso regulates imprinted DNA methylation of the H19 ICR in vivo. RESULTS Neither Kaiso deletion nor mutation of Kaiso binding sites in the 118-bp region affected DNA methylation of the mouse H19 ICR transgene. The endogenous mouse H19 ICR was methylated in a wild-type manner in Kaiso-null mutant mice. Additionally, the human IC1 transgene acquired imprinted DNA methylation after fertilization in the absence of Kaiso. CONCLUSIONS Our results indicate that Kaiso is not essential for either post-fertilization imprinted DNA methylation of the transgenic H19 ICR in mouse or for methylation imprinting of the endogenous mouse H19 ICR.
Collapse
Affiliation(s)
- Hitomi Matsuzaki
- Institute of Life and Environmental Sciences, Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Minami Kimura
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Mizuki Morihashi
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Keiji Tanimoto
- Institute of Life and Environmental Sciences, Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8577, Japan
| |
Collapse
|
43
|
Nakhod V, Krivenko A, Butkova T, Malsagova K, Kaysheva A. Advances in Molecular and Genetic Technologies and the Problems Related to Their Application in Personalized Medicine. J Pers Med 2024; 14:555. [PMID: 38929775 PMCID: PMC11204801 DOI: 10.3390/jpm14060555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 06/28/2024] Open
Abstract
Advances in the global personalized medicine market are directly related to innovations and developments in molecular and genetic technologies. This review focuses on the key trends in the development of these technologies in the healthcare sector. The existing global developments having an impact on the evolution of the personalized medicine market are reviewed. Efficient measures to support the development of molecular and genetic technologies are proposed.
Collapse
Affiliation(s)
- Valeriya Nakhod
- Institute of Biomedical Chemistry, 10 bld. 8, Pogodinskaya str., 119121 Moscow, Russia
| | | | | | | | | |
Collapse
|
44
|
Kanata E, Duffié R, Schulz EG. Establishment and maintenance of random monoallelic expression. Development 2024; 151:dev201741. [PMID: 38813842 PMCID: PMC11166465 DOI: 10.1242/dev.201741] [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] [Indexed: 05/31/2024]
Abstract
This Review elucidates the regulatory principles of random monoallelic expression by focusing on two well-studied examples: the X-chromosome inactivation regulator Xist and the olfactory receptor gene family. Although the choice of a single X chromosome or olfactory receptor occurs in different developmental contexts, common gene regulatory principles guide monoallelic expression in both systems. In both cases, an event breaks the symmetry between genetically and epigenetically identical copies of the gene, leading to the expression of one single random allele, stabilized through negative feedback control. Although many regulatory steps that govern the establishment and maintenance of monoallelic expression have been identified, key pieces of the puzzle are still missing. We provide an overview of the current knowledge and models for the monoallelic expression of Xist and olfactory receptors. We discuss their similarities and differences, and highlight open questions and approaches that could guide the study of other monoallelically expressed genes.
Collapse
Affiliation(s)
- Eleni Kanata
- Systems Epigenetics, Otto Warburg Laboratories, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Rachel Duffié
- Department of Biochemistry and Molecular Biophysics, Mortimer B. Zuckerman Mind, Brain, and Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Edda G. Schulz
- Systems Epigenetics, Otto Warburg Laboratories, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| |
Collapse
|
45
|
Niewold TB, Aksentijevich I, Gorevic PD, Gibson G, Yao Q. Genetically transitional disease: conceptual understanding and applicability to rheumatic disease. Nat Rev Rheumatol 2024; 20:301-310. [PMID: 38418715 DOI: 10.1038/s41584-024-01086-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 03/02/2024]
Abstract
In genomic medicine, the concept of genetically transitional disease (GTD) refers to cases in which gene mutation is necessary but not sufficient to cause disease. In this Perspective, we apply this novel concept to rheumatic diseases, which have been linked to hundreds of genetic variants via association studies. These variants are in the 'grey zone' between monogenic variants with large effect sizes and common susceptibility alleles with small effect sizes. Among genes associated with rare autoinflammatory diseases, many low-frequency and/or low-penetrance variants are known to increase susceptibility to systemic inflammation. In autoimmune diseases, hundreds of HLA and non-HLA genetic variants have been revealed to be modest- to moderate-risk alleles. These diseases can be reclassified as GTDs. The same concept could apply to many other human diseases. GTD could improve the reporting of genetic testing results, diagnostic yields, genetic counselling and selection of therapy, as well as facilitating research using a novel approach to human genetic diseases.
Collapse
Affiliation(s)
- Timothy B Niewold
- Department of Rheumatology, Hospital for Special Surgery, New York, NY, USA
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter D Gorevic
- Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Greg Gibson
- Center for Integrative Genomics, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Qingping Yao
- Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA.
| |
Collapse
|
46
|
Wei C, Kesner B, Yin H, Lee JT. Imprinted X chromosome inactivation at the gamete-to-embryo transition. Mol Cell 2024; 84:1442-1459.e7. [PMID: 38458200 PMCID: PMC11031340 DOI: 10.1016/j.molcel.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 12/23/2023] [Accepted: 02/13/2024] [Indexed: 03/10/2024]
Abstract
In mammals, dosage compensation involves two parallel processes: (1) X inactivation, which equalizes X chromosome dosage between males and females, and (2) X hyperactivation, which upregulates the active X for X-autosome balance. The field currently favors models whereby dosage compensation initiates "de novo" during mouse development. Here, we develop "So-Smart-seq" to revisit the question and interrogate a comprehensive transcriptome including noncoding genes and repeats in mice. Intriguingly, de novo silencing pertains only to a subset of Xp genes. Evolutionarily older genes and repetitive elements demonstrate constitutive Xp silencing, adopt distinct signatures, and do not require Xist to initiate silencing. We trace Xp silencing backward in developmental time to meiotic sex chromosome inactivation in the male germ line and observe that Xm hyperactivation is timed to Xp silencing on a gene-by-gene basis. Thus, during the gamete-to-embryo transition, older Xp genes are transmitted in a "pre-inactivated" state. These findings have implications for the evolution of imprinting.
Collapse
Affiliation(s)
- Chunyao Wei
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Barry Kesner
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Hao Yin
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Jeannie T Lee
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Giaccari C, Cecere F, Argenziano L, Pagano A, Galvao A, Acampora D, Rossi G, Hay Mele B, Acurzio B, Coonrod S, Cubellis MV, Cerrato F, Andrews S, Cecconi S, Kelsey G, Riccio A. A maternal-effect Padi6 variant causes nuclear and cytoplasmic abnormalities in oocytes, as well as failure of epigenetic reprogramming and zygotic genome activation in embryos. Genes Dev 2024; 38:131-150. [PMID: 38453481 PMCID: PMC10982689 DOI: 10.1101/gad.351238.123] [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: 10/09/2023] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Maternal inactivation of genes encoding components of the subcortical maternal complex (SCMC) and its associated member, PADI6, generally results in early embryo lethality. In humans, SCMC gene variants were found in the healthy mothers of children affected by multilocus imprinting disturbances (MLID). However, how the SCMC controls the DNA methylation required to regulate imprinting remains poorly defined. We generated a mouse line carrying a Padi6 missense variant that was identified in a family with Beckwith-Wiedemann syndrome and MLID. If homozygous in female mice, this variant resulted in interruption of embryo development at the two-cell stage. Single-cell multiomic analyses demonstrated defective maturation of Padi6 mutant oocytes and incomplete DNA demethylation, down-regulation of zygotic genome activation (ZGA) genes, up-regulation of maternal decay genes, and developmental delay in two-cell embryos developing from Padi6 mutant oocytes but little effect on genomic imprinting. Western blotting and immunofluorescence analyses showed reduced levels of UHRF1 in oocytes and abnormal localization of DNMT1 and UHRF1 in both oocytes and zygotes. Treatment with 5-azacytidine reverted DNA hypermethylation but did not rescue the developmental arrest of mutant embryos. Taken together, this study demonstrates that PADI6 controls both nuclear and cytoplasmic oocyte processes that are necessary for preimplantation epigenetic reprogramming and ZGA.
Collapse
Affiliation(s)
- Carlo Giaccari
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania "Luigi Vanvitelli," Caserta 81100, Italy
| | - Francesco Cecere
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania "Luigi Vanvitelli," Caserta 81100, Italy
| | - Lucia Argenziano
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania "Luigi Vanvitelli," Caserta 81100, Italy
| | - Angela Pagano
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania "Luigi Vanvitelli," Caserta 81100, Italy
| | - Antonio Galvao
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, United Kingdom
- Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn 10-748, Poland
| | - Dario Acampora
- Institute of Genetics and Biophysics (IGB) "Adriano Buzzati-Traverso," Consiglio Nazionale delle Ricerche (CNR), Naples 80131, Italy
| | - Gianna Rossi
- Department of Life, Health, and Environmental Sciences, Università dell'Aquila, L'Aquila 67100, Italy
| | - Bruno Hay Mele
- Department of Biology, University of Naples "Federico II," Napoli 80126, Italy
| | - Basilia Acurzio
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania "Luigi Vanvitelli," Caserta 81100, Italy
| | - Scott Coonrod
- Baker Institute for Animal Health, Cornell University, Ithaca, New York 14853, USA
| | | | - Flavia Cerrato
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania "Luigi Vanvitelli," Caserta 81100, Italy
| | - Simon Andrews
- Bioinformatics Unit, The Babraham Institute, Cambridge CB22 3AT, United Kingdom
| | - Sandra Cecconi
- Department of Life, Health, and Environmental Sciences, Università dell'Aquila, L'Aquila 67100, Italy
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, United Kingdom;
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, United Kingdom
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Cambridge CB2 0QQ, United Kingdom
| | - Andrea Riccio
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania "Luigi Vanvitelli," Caserta 81100, Italy;
- Institute of Genetics and Biophysics (IGB) "Adriano Buzzati-Traverso," Consiglio Nazionale delle Ricerche (CNR), Naples 80131, Italy
| |
Collapse
|
48
|
Lin Y, Li J, Gu Y, Jin L, Bai J, Zhang J, Wang Y, Liu P, Long K, He M, Li D, Liu C, Han Z, Zhang Y, Li X, Zeng B, Lu L, Kong F, Sun Y, Fan Y, Wang X, Wang T, Jiang A, Ma J, Shen L, Zhu L, Jiang Y, Tang G, Fan X, Liu Q, Li H, Wang J, Chen L, Ge L, Li X, Tang Q, Li M. Haplotype-resolved 3D chromatin architecture of the hybrid pig. Genome Res 2024; 34:310-325. [PMID: 38479837 PMCID: PMC10984390 DOI: 10.1101/gr.278101.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024]
Abstract
In diploid mammals, allele-specific three-dimensional (3D) genome architecture may lead to imbalanced gene expression. Through ultradeep in situ Hi-C sequencing of three representative somatic tissues (liver, skeletal muscle, and brain) from hybrid pigs generated by reciprocal crosses of phenotypically and physiologically divergent Berkshire and Tibetan pigs, we uncover extensive chromatin reorganization between homologous chromosomes across multiple scales. Haplotype-based interrogation of multi-omic data revealed the tissue dependence of 3D chromatin conformation, suggesting that parent-of-origin-specific conformation may drive gene imprinting. We quantify the effects of genetic variations and histone modifications on allelic differences of long-range promoter-enhancer contacts, which likely contribute to the phenotypic differences between the parental pig breeds. We also observe the fine structure of somatically paired homologous chromosomes in the pig genome, which has a functional implication genome-wide. This work illustrates how allele-specific chromatin architecture facilitates concomitant shifts in allele-biased gene expression, as well as the possible consequential phenotypic changes in mammals.
Collapse
Affiliation(s)
- Yu Lin
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jing Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
| | - Yiren Gu
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Long Jin
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jingyi Bai
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaman Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yujie Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Pengliang Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Keren Long
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Mengnan He
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Diyan Li
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Can Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Ziyin Han
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yu Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaokai Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Bo Zeng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Lu Lu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Fanli Kong
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Ying Sun
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Institute of Geriatric Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yongliang Fan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xun Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Wang
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - An'an Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jideng Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Linyuan Shen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yanzhi Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Guoqing Tang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaolan Fan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Qingyou Liu
- Animal Molecular Design and Precise Breeding Key Laboratory of Guangdong Province, School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Hua Li
- Animal Molecular Design and Precise Breeding Key Laboratory of Guangdong Province, School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Jinyong Wang
- Pig Industry Sciences Key Laboratory of Ministry of Agriculture and Rural Affairs, Chongqing Academy of Animal Sciences, Chongqing 402460, China
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Li Chen
- Pig Industry Sciences Key Laboratory of Ministry of Agriculture and Rural Affairs, Chongqing Academy of Animal Sciences, Chongqing 402460, China
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Liangpeng Ge
- Pig Industry Sciences Key Laboratory of Ministry of Agriculture and Rural Affairs, Chongqing Academy of Animal Sciences, Chongqing 402460, China
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Xuewei Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Qianzi Tang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
| |
Collapse
|
49
|
Yuan S, Gao L, Tao W, Zhan J, Lu G, Zhang J, Zhang C, Yi L, Liu Z, Hou Z, Dai M, Zhao H, Chen ZJ, Liu J, Wu K. Allelic reprogramming of chromatin states in human early embryos. Natl Sci Rev 2024; 11:nwad328. [PMID: 38449877 PMCID: PMC10917445 DOI: 10.1093/nsr/nwad328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/04/2023] [Accepted: 12/16/2023] [Indexed: 03/08/2024] Open
Abstract
The reprogramming of parental epigenomes in human early embryos remains elusive. To what extent the characteristics of parental epigenomes are conserved between humans and mice is currently unknown. Here, we mapped parental haploid epigenomes using human parthenogenetic and androgenetic embryos. Human embryos have a larger portion of genome with parentally specific epigenetic states than mouse embryos. The allelic patterns of epigenetic states for orthologous regions are not conserved between humans and mice. Nevertheless, it is conserved that maternal DNA methylation and paternal H3K27me3 are associated with the repression of two alleles in humans and mice. In addition, for DNA-methylation-dependent imprinting, we report 19 novel imprinted genes and their associated germline differentially methylated regions. Unlike in mice, H3K27me3-dependent imprinting is not observed in human early embryos. Collectively, allele-specific epigenomic reprogramming is different in humans and mice.
Collapse
Affiliation(s)
- Shenli Yuan
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China
- CAS Key Laboratory of Genome Sciences and Information, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Lei Gao
- CAS Key Laboratory of Genome Sciences and Information, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
| | - Wenrong Tao
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Reproductive Endocrinology of the Ministry of Education, Shandong University, Jinan 250012, China
| | - Jianhong Zhan
- CAS Key Laboratory of Genome Sciences and Information, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Jingye Zhang
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Reproductive Endocrinology of the Ministry of Education, Shandong University, Jinan 250012, China
| | - Chuanxin Zhang
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Reproductive Endocrinology of the Ministry of Education, Shandong University, Jinan 250012, China
| | - Lizhi Yi
- CAS Key Laboratory of Genome Sciences and Information, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenbo Liu
- CAS Key Laboratory of Genome Sciences and Information, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenzhen Hou
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Reproductive Endocrinology of the Ministry of Education, Shandong University, Jinan 250012, China
| | - Min Dai
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Reproductive Endocrinology of the Ministry of Education, Shandong University, Jinan 250012, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Reproductive Endocrinology of the Ministry of Education, Shandong University, Jinan 250012, China
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Jiang Liu
- CAS Key Laboratory of Genome Sciences and Information, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Keliang Wu
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Reproductive Endocrinology of the Ministry of Education, Shandong University, Jinan 250012, China
| |
Collapse
|
50
|
Dong X, Luo H, Yao J, Guo Q, Yu S, Ruan Y, Li F, Jin W, Meng D. The conservation of allelic DNA methylation and its relationship with imprinting in maize. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:1376-1389. [PMID: 37935439 PMCID: PMC10901201 DOI: 10.1093/jxb/erad440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023]
Abstract
Genomic imprinting refers to allele-specific expression of genes depending on parental origin, and it is regulated by epigenetic modifications. Intraspecific allelic variation for imprinting has been detected; however, the intraspecific genome-wide allelic epigenetic variation in maize and its correlation with imprinting variants remain unclear. Here, three reciprocal hybrids were generated by crossing Zea mays inbred lines CAU5, B73, and Mo17 in order to examine the intraspecific conservation of the imprinted genes in the kernel. The majority of imprinted genes exhibited intraspecific conservation, and these genes also exhibited interspecific conservation (rice, sorghum, and Arabidopsis) and were enriched in some specific pathways. By comparing intraspecific allelic DNA methylation in the endosperm, we found that nearly 15% of DNA methylation existed as allelic variants. The intraspecific whole-genome correlation between DNA methylation and imprinted genes indicated that DNA methylation variants play an important role in imprinting variants. Disruption of two conserved imprinted genes using CRISPR/Cas9 editing resulted in a smaller kernel phenotype. Our results shed light on the intraspecific correlation of DNA methylation variants and variation for imprinting in maize, and show that imprinted genes play an important role in kernel development.
Collapse
Affiliation(s)
- Xiaomei Dong
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, Liaoning, China
- Shenyang City Key Laboratory of Maize Genomic Selection Breeding, Shenyang 110866, Liaoning, China
| | - Haishan Luo
- College of Agronomy, Shenyang Agricultural University, Shenyang 110866, Liaoning, China
| | - Jiabin Yao
- College of Agronomy, Shenyang Agricultural University, Shenyang 110866, Liaoning, China
| | - Qingfeng Guo
- College of Agronomy, Shenyang Agricultural University, Shenyang 110866, Liaoning, China
| | - Shuai Yu
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, Liaoning, China
- Shenyang City Key Laboratory of Maize Genomic Selection Breeding, Shenyang 110866, Liaoning, China
| | - Yanye Ruan
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, Liaoning, China
- Shenyang City Key Laboratory of Maize Genomic Selection Breeding, Shenyang 110866, Liaoning, China
| | - Fenghai Li
- College of Agronomy, Shenyang Agricultural University, Shenyang 110866, Liaoning, China
| | - Weiwei Jin
- State Key Laboratory of Plant Physiology and Biochemistry, National Maize Improvement Center, Key Laboratory of Crop Heterosis and Utilization, the Ministry of Education, College of Agronomy and Biotechnology, China Agricultural University, Beijing 100193, China
- Department of Agronomy, College of Agriculture & Resources and Environmental Sciences, Tianjin Agricultural University, Tianjin 300392, China
| | - Dexuan Meng
- College of Agronomy, Shenyang Agricultural University, Shenyang 110866, Liaoning, China
| |
Collapse
|