Impaired Reorganization of Centrosome Structure Underlies Human Infantile Dilated Cardiomyopathy

Background: During cardiomyocyte maturation, the centrosome, which functions as a microtubule organizing center in cardiomyocytes, undergoes dramatic structural reorganization where its components reorganize from being localized at the centriole to the nuclear envelope. This developmentally programmed process, referred to as centrosome reduction, has been previously associated with cell cycle exit. However, understanding of how this process influences cardiomyocyte cell biology, and whether its disruption results in human cardiac disease, remains unknown. We studied this phenomenon in an infant with a rare case of infantile dilated cardiomyopathy (iDCM) who presented with left ventricular ejection fraction of 18% and disrupted sarcomere and mitochondria structure. Methods: We performed an analysis beginning with an infant who presented with a rare case of iDCM. We derived induced pluripotent stem cells from the patient to model iDCM in vitro. We performed whole exome sequencing on the patient and his parents for causal gene analysis. CRISPR/Cas9-mediated gene knockout and correction in vitro were used to confirm whole exome sequencing results. Zebrafish and Drosophila models were used for in vivo validation of the causal gene. Matrigel mattress technology and single-cell RNA sequencing were used to characterize iDCM cardiomyocytes further. Results: Whole exome sequencing and CRISPR/Cas9 gene knockout/correction identified RTTN, the gene encoding the centrosomal protein RTTN (rotatin), as the causal gene underlying the patient’s condition, representing the first time a centrosome defect has been implicated in a nonsyndromic dilated cardiomyopathy. Genetic knockdowns in zebrafish and Drosophila confirmed an evolutionarily conserved requirement of RTTN for cardiac structure and function. Single-cell RNA sequencing of iDCM cardiomyocytes showed impaired maturation of iDCM cardiomyocytes, which underlie the observed cardiomyocyte structural and functional deficits. We also observed persistent localization of the centrosome at the centriole, contrasting with expected programmed perinuclear reorganization, which led to subsequent global microtubule network defects. In addition, we identified a small molecule that restored centrosome reorganization and improved the structure and contractility of iDCM cardiomyocytes. Conclusions: This study is the first to demonstrate a case of human disease caused by a defect in centrosome reduction. We also uncovered a novel role for RTTN in perinatal cardiac development and identified a potential therapeutic strategy for centrosome-related iDCM. Future study aimed at identifying variants in centrosome components may uncover additional contributors to human cardiac disease.

[1]  I. Konstantinov,et al.  Defining the Fetal Gene Program at Single-Cell Resolution in Pediatric Dilated Cardiomyopathy , 2022, Circulation.

[2]  N. Blom,et al.  Genetic Evaluation of A Nation-Wide Dutch Pediatric DCM Cohort: The Use of Genetic Testing in Risk Stratification , 2022, Circulation. Genomic and precision medicine.

[3]  K. Yap,et al.  Genotype and Cardiac Outcomes in Pediatric Dilated Cardiomyopathy , 2021, Journal of the American Heart Association.

[4]  Oriol Vinyals,et al.  Highly accurate protein structure prediction with AlphaFold , 2021, Nature.

[5]  Silvia Vergarajauregui,et al.  Alternative Splicing of Pericentrin Contributes to Cell Cycle Control in Cardiomyocytes , 2021, bioRxiv.

[6]  Matthew Miyamoto,et al.  Heart organoids and tissue models for modeling development and disease. , 2021, Seminars in cell & developmental biology.

[7]  J. Pagan,et al.  Centrosome Reduction Promotes Terminal Differentiation of Human Cardiomyocytes , 2020, Stem cell reports.

[8]  Marina Leone,et al.  Microtubule Organization in Striated Muscle Cells , 2020, Cells.

[9]  Matthew Miyamoto,et al.  Transcriptomic entropy benchmarks stem cell-derived cardiomyocyte maturation against endogenous tissue at single cell level , 2020, bioRxiv.

[10]  Hideki Uosaki,et al.  A Novel Fluorescent Reporter System Identifies Laminin-511/521 as Potent Regulators of Cardiomyocyte Maturation , 2020, Scientific Reports.

[11]  M. Aschner,et al.  Huntington's Disease associated resistance to Mn neurotoxicity is neurodevelopmental stage and neuronal lineage dependent. , 2019, Neurotoxicology.

[12]  D. Roden,et al.  Real-time visualization of titin dynamics reveals extensive reversible photobleaching in human induced pluripotent stem cell-derived cardiomyocytes. , 2019, American journal of physiology. Cell physiology.

[13]  M. Giacca,et al.  Cardiac regeneration and remodelling of the cardiomyocyte cytoarchitecture , 2019, The FEBS journal.

[14]  S. Colan,et al.  Cardiomyopathy in Children: Classification and Diagnosis: A Scientific Statement From the American Heart Association. , 2019, Circulation.

[15]  S. Baig,et al.  Primary microcephaly, primordial dwarfism, and brachydactyly in adult cases with biallelic skipping of RTTN exon 42 , 2019, Human mutation.

[16]  G. Schaaf,et al.  Heterogeneous clinical phenotypes and cerebral malformations reflected by rotatin cellular dynamics , 2019, Brain : a journal of neurology.

[17]  R. Satija,et al.  Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression , 2019, Genome Biology.

[18]  Guang Li,et al.  Single cell expression analysis reveals anatomical and cell cycle-dependent transcriptional shifts during heart development , 2019, Development.

[19]  S. Seneca,et al.  Biallelic mutations in RTTN are associated with microcephaly, short stature and a wide range of brain malformations. , 2018, European journal of medical genetics.

[20]  N. Boddaert,et al.  Recurrent RTTN mutation leading to severe microcephaly, polymicrogyria and growth restriction. , 2018, European journal of medical genetics.

[21]  The Gene Ontology Consortium,et al.  The Gene Ontology Resource: 20 years and still GOing strong , 2018, Nucleic Acids Res..

[22]  C. Vasilescu,et al.  Genetic Basis of Severe Childhood-Onset Cardiomyopathies. , 2018, Journal of the American College of Cardiology.

[23]  B. Raught,et al.  PPP1R35 is a novel centrosomal protein that regulates centriole length in concert with the microcephaly protein RTTN , 2018, eLife.

[24]  J. Shimony,et al.  Functional Characterization of Biallelic RTTN Variants Identified in an Infant with Microcephaly, Simplified Gyral Pattern, Pontocerebellar Hypoplasia, and Seizures , 2018, Pediatric Research.

[25]  Paul Hoffman,et al.  Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.

[26]  R. Sinke,et al.  Toward an effective exome-based genetic testing strategy in pediatric dilated cardiomyopathy , 2018, Genetics in Medicine.

[27]  K. He,et al.  Mitochondria are transported along microtubules in membrane nanotubes to rescue distressed cardiomyocytes from apoptosis , 2018, Cell Death & Disease.

[28]  A. Muroyama,et al.  Microtubule organization, dynamics and functions in differentiated cells , 2017, Development.

[29]  T. Tang,et al.  Human microcephaly protein RTTN interacts with STIL and is required to build full-length centrioles , 2017, Nature Communications.

[30]  Jared Evans,et al.  Diagnostic Yield of Whole Exome Sequencing in Pediatric Dilated Cardiomyopathy , 2017, Journal of cardiovascular development and disease.

[31]  C. Santoro,et al.  Expanding the phenotype of RTTN variations: a new family with primary microcephaly, severe growth failure, brain malformations and dermatitis , 2016, Clinical genetics.

[32]  Kari Naylor,et al.  Microtubules Are Essential for Mitochondrial Dynamics–Fission, Fusion, and Motility–in Dictyostelium discoideum , 2016, Front. Cell Dev. Biol..

[33]  M. Aschner,et al.  Genomic Instability Associated with p53 Knockdown in the Generation of Huntington’s Disease Human Induced Pluripotent Stem Cells , 2016, PloS one.

[34]  F. Alkuraya,et al.  RTTN Mutations Cause Primary Microcephaly and Primordial Dwarfism in Humans. , 2015, American journal of human genetics.

[35]  C. Hong,et al.  Combinatorial polymer matrices enhance in vitro maturation of human induced pluripotent stem cell-derived cardiomyocytes. , 2015, Biomaterials.

[36]  Jonathan E. Hempel,et al.  Matrigel Mattress: A Method for the Generation of Single Contracting Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes. , 2015, Circulation research.

[37]  D. Zebrowski,et al.  Developmental alterations in centrosome integrity contribute to the post-mitotic state of mammalian cardiomyocytes , 2015, eLife.

[38]  Yan Guo,et al.  Three-stage quality control strategies for DNA re-sequencing data , 2014, Briefings Bioinform..

[39]  Jiang Li,et al.  Multi-perspective quality control of Illumina exome sequencing data using QC3. , 2014, Genomics.

[40]  A. Rebbaa,et al.  Identification, Mechanism of Action, and Antitumor Activity of a Small Molecule Inhibitor of Hippo, TGF-β, and Wnt Signaling Pathways , 2014, Molecular Cancer Therapeutics.

[41]  L. Fernandez,et al.  Mutations in Alström Protein Impair Terminal Differentiation of Cardiomyocytes , 2014, Nature Communications.

[42]  A. Engler,et al.  A Drosophila melanogaster Model of Diastolic Dysfunction and Cardiomyopathy Based on Impaired Troponin-T Function , 2014, Circulation research.

[43]  Feng Zhang,et al.  Genome engineering using CRISPR-Cas9 system. , 2015, Methods in molecular biology.

[44]  Min Wu,et al.  Structural and biomechanical basis of mitochondrial movement in eukaryotic cells , 2013, International journal of nanomedicine.

[45]  Luke A. Gilbert,et al.  CRISPR-Mediated Modular RNA-Guided Regulation of Transcription in Eukaryotes , 2013, Cell.

[46]  Sean P. Palecek,et al.  Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions , 2012, Nature Protocols.

[47]  Erik Engelen,et al.  RTTN mutations link primary cilia function to organization of the human cerebral cortex. , 2012, American journal of human genetics.

[48]  Nicole O Glenn,et al.  The W-Loop of Alpha-Cardiac Actin Is Critical for Heart Function and Endocardial Cushion Morphogenesis in Zebrafish , 2012, Molecular and Cellular Biology.

[49]  Johannes E. Schindelin,et al.  Fiji: an open-source platform for biological-image analysis , 2012, Nature Methods.

[50]  Xiaolei Xu,et al.  Immunostaining of dissected zebrafish embryonic heart. , 2012, Journal of visualized experiments : JoVE.

[51]  M. DePristo,et al.  A framework for variation discovery and genotyping using next-generation DNA sequencing data , 2011, Nature Genetics.

[52]  H. Hakonarson,et al.  ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data , 2010, Nucleic acids research.

[53]  C. Wurzenberger,et al.  Jcb: Report , 2022 .

[54]  Wei Huang,et al.  Myofibrillogenesis in the developing zebrafish heart: A functional study of tnnt2. , 2009, Developmental biology.

[55]  E. Olson,et al.  Hand, an evolutionarily conserved bHLH transcription factor required for Drosophila cardiogenesis and hematopoiesis , 2006, Development.

[56]  Joseph A Izatt,et al.  Drosophila as a model for the identification of genes causing adult human heart disease , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[57]  E. Olson,et al.  Hand is a direct target of Tinman and GATA factors during Drosophila cardiogenesis and hematopoiesis , 2005, Development.

[58]  P. Gruss,et al.  Rotatin is a novel gene required for axial rotation and left–right specification in mouse embryos , 2002, Mechanisms of Development.

[59]  M. Ashburner,et al.  Gene Ontology: tool for the unification of biology , 2000, Nature Genetics.

[60]  R Plomin,et al.  A simple method for analyzing microsatellite allele image patterns generated from DNA pools and its application to allelic association studies. , 1998, American journal of human genetics.

[61]  C. Kimmel,et al.  Stages of embryonic development of the zebrafish , 1995, Developmental dynamics : an official publication of the American Association of Anatomists.

[62]  W. Chung,et al.  Pediatric Cardiomyopathies. , 2017, Circulation research.