Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 625
Filter
Add more filters

Publication year range
1.
Annu Rev Cell Dev Biol ; 35: 131-168, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31399000

ABSTRACT

Protein coats are supramolecular complexes that assemble on the cytosolic face of membranes to promote cargo sorting and transport carrier formation in the endomembrane system of eukaryotic cells. Several types of protein coats have been described, including COPI, COPII, AP-1, AP-2, AP-3, AP-4, AP-5, and retromer, which operate at different stages of the endomembrane system. Defects in these coats impair specific transport pathways, compromising the function and viability of the cells. In humans, mutations in subunits of these coats cause various congenital diseases that are collectively referred to as coatopathies. In this article, we review the fundamental properties of protein coats and the diseases that result from mutation of their constituent subunits.


Subject(s)
Endosomes/chemistry , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Vesicular Transport Proteins/genetics , Animals , Coat Protein Complex I/genetics , Coat Protein Complex I/metabolism , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/therapy , Humans , Protein Transport , Vesicular Transport Proteins/metabolism
2.
Nat Rev Mol Cell Biol ; 20(12): 766-784, 2019 12.
Article in English | MEDLINE | ID: mdl-31558824

ABSTRACT

The spatiotemporal control of RNA polymerase II (Pol II)-mediated gene transcription is tightly and intricately regulated. In addition, preservation of the integrity of the DNA template is required so as to ensure unperturbed transcription, particularly since DNA is continually challenged by different types of damaging agents that can form transcription-blocking DNA lesions (TBLs), which impede transcription elongation and cause transcription stress. To overcome the highly cytotoxic effects of TBLs, an intricate cellular response has evolved, in which the transcription-coupled nucleotide excision repair (TC-NER) pathway has a central role in removing TBLs specifically from the transcribed strand. Damage detection by stalling of the transcribing Pol II is highly efficient, but a stalled Pol II complex may create an even bigger problem by interfering with repair of the lesions, and overall with transcription and replication. In this Review, we discuss the effects of different types of DNA damage on Pol II, important concepts of transcription stress, the manner in which TBLs are removed by TC-NER and how different tissues respond to TBLs. We also discuss the role of TBLs in ageing and the complex genotype-phenotype correlations of TC-NER hereditary disorders.


Subject(s)
DNA Damage , DNA Repair , DNA Replication , Genetic Diseases, Inborn , RNA Polymerase II/metabolism , Transcription, Genetic , Animals , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , RNA Polymerase II/genetics
3.
Nat Rev Mol Cell Biol ; 18(12): 771-783, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28974772

ABSTRACT

Cellular mechanotransduction, the process of translating mechanical forces into biological signals, is crucial for a wide range of physiological processes. A role for ion channels in sensing mechanical forces has been proposed for decades, but their identity in mammals remained largely elusive until the discovery of Piezos. Recent research on Piezos has underscored their importance in somatosensation (touch perception, proprioception and pulmonary respiration), red blood cell volume regulation, vascular physiology and various human genetic disorders.


Subject(s)
Genetic Diseases, Inborn/metabolism , Ion Channel Gating , Ion Channels/metabolism , Proprioception , Respiratory Mechanics , Touch Perception , Animals , Genetic Diseases, Inborn/genetics , Humans , Ion Channels/genetics
4.
Nat Rev Mol Cell Biol ; 17(3): 170-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26818440

ABSTRACT

Experimental modelling of human disorders enables the definition of the cellular and molecular mechanisms underlying diseases and the development of therapies for treating them. The availability of human pluripotent stem cells (PSCs), which are capable of self-renewal and have the potential to differentiate into virtually any cell type, can now help to overcome the limitations of animal models for certain disorders. The ability to model human diseases using cultured PSCs has revolutionized the ways in which we study monogenic, complex and epigenetic disorders, as well as early- and late-onset diseases. Several strategies are used to generate such disease models using either embryonic stem cells (ES cells) or patient-specific induced PSCs (iPSCs), creating new possibilities for the establishment of models and their use in drug screening.


Subject(s)
Genetic Diseases, Inborn , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Stem Cell Transplantation/methods , Allografts , Animals , Autografts , Disease Models, Animal , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/therapy , Humans
5.
Hum Mol Genet ; 32(9): 1457-1465, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36458889

ABSTRACT

Mosaic variants in the PIK3CA gene, encoding the catalytic subunit of phosphoinositide 3-kinase (PI3K), produce constitutive PI3K activation, which causes PIK3CA-related overgrowth spectrum disorders. To date, fewer than 20 patients have been described with germline alterations in PIK3CA. In this study, we describe three unrelated individuals with overgrowth and germline PIK3CA variants. These variants were discovered through whole-exome sequencing and confirmed as germline by testing multiple tissue types, when available. Functional analysis using Patient 1's fibroblast cell line and two previously reported patients' cell lines showed increased phosphorylation of AKT during cellular starvation revealing constitutive activation of the phosphoinositide-3-kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR) pathway. Alternatively, stimulation of the cells by fetal bovine serum produced a reduced response, indicating an activated status of the PI3K complex reducing the pathway response to further external stimulation. Additional studies utilizing Biolog Phenotype Microarray technology indicated reduced energy production when cells were exposed to growth factors stimulating the PI3K/AKT/mTOR pathway, confirming the trend observed in the AKT phosphorylation test after stimulation. Furthermore, treatment with inhibitors of the PI3K/AKT/mTOR pathway rescued the normal energy response in the patients' cells. Collectively, these data demonstrate that disease-causing germline PIK3CA variants have a functional consequence, similar to mosaic variants in the PI3K/AKT/mTOR pathway.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Genetic Diseases, Inborn , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Germ Cells/metabolism , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/physiopathology , Germ-Line Mutation , Phosphorylation
6.
Am J Hum Genet ; 109(2): 210-222, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35065709

ABSTRACT

Variable levels of gene expression between tissues complicates the use of RNA sequencing of patient biosamples to delineate the impact of genomic variants. Here, we describe a gene- and tissue-specific metric to inform the feasibility of RNA sequencing. This overcomes limitations of using expression values alone as a metric to predict RNA-sequencing utility. We have derived a metric, minimum required sequencing depth (MRSD), that estimates the depth of sequencing required from RNA sequencing to achieve user-specified sequencing coverage of a gene, transcript, or group of genes. We applied MRSD across four human biosamples: whole blood, lymphoblastoid cell lines (LCLs), skeletal muscle, and cultured fibroblasts. MRSD has high precision (90.1%-98.2%) and overcomes transcript region-specific sequencing biases. Applying MRSD scoring to established disease gene panels shows that fibroblasts, of these four biosamples, are the optimum source of RNA for 63.1% of gene panels. Using this approach, up to 67.8% of the variants of uncertain significance in ClinVar that are predicted to impact splicing could be assayed by RNA sequencing in at least one of the biosamples. We demonstrate the utility and benefits of MRSD as a metric to inform functional assessment of splicing aberrations, in particular in the context of Mendelian genetic disorders to improve diagnostic yield.


Subject(s)
Genetic Diseases, Inborn/genetics , RNA Splicing , RNA, Messenger/genetics , Sequence Analysis, RNA/statistics & numerical data , Software , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Blood Cells/metabolism , Blood Cells/pathology , Cell Line , Fibroblasts/metabolism , Fibroblasts/pathology , Genetic Diseases, Inborn/classification , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Genetic Variation , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , RNA, Messenger/metabolism , Research Design , Exome Sequencing/statistics & numerical data
7.
Mol Genet Metab ; 142(1): 108360, 2024 May.
Article in English | MEDLINE | ID: mdl-38428378

ABSTRACT

The Mendelian disorders of chromatin machinery (MDCMs) represent a distinct subgroup of disorders that present with neurodevelopmental disability. The chromatin machinery regulates gene expression by a range of mechanisms, including by post-translational modification of histones, responding to histone marks, and remodelling nucleosomes. Some of the MDCMs that impact on histone modification may have potential therapeutic interventions. Two potential treatment strategies are to enhance the intracellular pool of metabolites that can act as substrates for histone modifiers and the use of medications that may inhibit or promote the modification of histone residues to influence gene expression. In this article we discuss the influence and potential treatments of histone modifications involving histone acetylation and histone methylation. Genomic technologies are facilitating earlier diagnosis of many Mendelian disorders, providing potential opportunities for early treatment from infancy. This has parallels with how inborn errors of metabolism have been afforded early treatment with newborn screening. Before this promise can be fulfilled, we require greater understanding of the biochemical fingerprint of these conditions, which may provide opportunities to supplement metabolites that can act as substrates for chromatin modifying enzymes. Importantly, understanding the metabolomic profile of affected individuals may also provide disorder-specific biomarkers that will be critical for demonstrating efficacy of treatment, as treatment response may not be able to be accurately assessed by clinical measures.


Subject(s)
Chromatin , Metabolic Networks and Pathways , Humans , Chromatin/genetics , Chromatin/metabolism , Metabolic Networks and Pathways/genetics , Histones/metabolism , Histones/genetics , Protein Processing, Post-Translational , Acetylation , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/therapy , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/metabolism , Chromatin Assembly and Disassembly/genetics , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Genetic Diseases, Inborn/metabolism , Infant, Newborn , Methylation
8.
PLoS Biol ; 19(4): e3001207, 2021 04.
Article in English | MEDLINE | ID: mdl-33909605

ABSTRACT

Missense variants are present amongst the healthy population, but some of them are causative of human diseases. A classification of variants associated with "healthy" or "diseased" states is therefore not always straightforward. A deeper understanding of the nature of missense variants in health and disease, the cellular processes they may affect, and the general molecular principles which underlie these differences is essential to offer mechanistic explanations of the true impact of pathogenic variants. Here, we have formalised a statistical framework which enables robust probabilistic quantification of variant enrichment across full-length proteins, their domains, and 3D structure-defined regions. Using this framework, we validate and extend previously reported trends of variant enrichment in different protein structural regions (surface/core/interface). By examining the association of variant enrichment with available functional pathways and transcriptomic and proteomic (protein half-life, thermal stability, abundance) data, we have mined a rich set of molecular features which distinguish between pathogenic and population variants: Pathogenic variants mainly affect proteins involved in cell proliferation and nucleotide processing and are enriched in more abundant proteins. Additionally, rare population variants display features closer to common than pathogenic variants. We validate the association between these molecular features and variant pathogenicity by comparing against existing in silico variant impact annotations. This study provides molecular details into how different proteins exhibit resilience and/or sensitivity towards missense variants and provides the rationale to prioritise variant-enriched proteins and protein domains for therapeutic targeting and development. The ZoomVar database, which we created for this study, is available at fraternalilab.kcl.ac.uk/ZoomVar. It allows users to programmatically annotate missense variants with protein structural information and to calculate variant enrichment in different protein structural regions.


Subject(s)
Genetic Diseases, Inborn/genetics , Mutation, Missense/physiology , Proteome , Amino Acid Sequence/genetics , Computational Biology/methods , Databases, Protein , Genetic Diseases, Inborn/metabolism , Genetic Predisposition to Disease , Germ-Line Mutation , Health , Humans , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Conformation , Protein Folding , Protein Interaction Domains and Motifs/genetics , Protein Processing, Post-Translational/genetics , Proteins/genetics , Proteins/metabolism , Proteome/chemistry , Proteome/genetics , Proteome/metabolism , Proteomics , Signal Transduction/genetics , Software
9.
Hum Mol Genet ; 30(23): 2215-2224, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34230955

ABSTRACT

CHARGE syndrome is an autosomal dominant malformation disorder caused by pathogenic variants in the chromatin remodeler CHD7. Affected are craniofacial structures, cranial nerves and multiple organ systems. Depending on the combination of malformations present, its distinction from other congenital disorders can be challenging. To gain a better insight into the regulatory disturbances in CHARGE syndrome, we performed RNA-Seq analysis on blood samples of 19 children with CHARGE syndrome and a confirmed disease-causing CHD7 variant in comparison with healthy control children. Our analysis revealed a distinct CHARGE syndrome pattern with downregulation of genes that are linked to disorders described to mimic the CHARGE phenotype, i.e. KMT2D and KDM6A (Kabuki syndrome), EP300 and CREBBP (Rubinstein-Taybi syndrome) and ARID1A and ARID1B (Coffin-Siris syndrome). Furthermore, by performing protein-protein interaction studies using co-immunoprecipitation, direct yeast-two hybrid and in situ proximity ligation assays, we could demonstrate an interplay between CHD7, KMT2D, KDM6A and EP300. In summary, our data demonstrate a mechanistic and regulatory link between the developmental disorders CHARGE-, Kabuki- and Rubinstein Taybi-syndrome providing an explanation for the overlapping phenotypes.


Subject(s)
CHARGE Syndrome/diagnosis , CHARGE Syndrome/genetics , Genetic Association Studies , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease , Age Factors , CHARGE Syndrome/metabolism , Carrier Proteins , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Genetic Association Studies/methods , Genetic Diseases, Inborn/metabolism , Genetic Markers , Genetic Variation , Humans , Immunoprecipitation , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phenotype , Protein Binding , RNA-Seq
10.
Nucleic Acids Res ; 49(1): 53-66, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33300042

ABSTRACT

Assessing the causal tissues of human complex diseases is important for the prioritization of trait-associated genetic variants. Yet, the biological underpinnings of trait-associated variants are extremely difficult to infer due to statistical noise in genome-wide association studies (GWAS), and because >90% of genetic variants from GWAS are located in non-coding regions. Here, we collected the largest human epigenomic map from ENCODE and Roadmap consortia and implemented a deep-learning-based convolutional neural network (CNN) model to predict the regulatory roles of genetic variants across a comprehensive list of epigenomic modifications. Our model, called DeepFun, was built on DNA accessibility maps, histone modification marks, and transcription factors. DeepFun can systematically assess the impact of non-coding variants in the most functional elements with tissue or cell-type specificity, even for rare variants or de novo mutations. By applying this model, we prioritized trait-associated loci for 51 publicly-available GWAS studies. We demonstrated that CNN-based analyses on dense and high-resolution epigenomic annotations can refine important GWAS associations in order to identify regulatory loci from background signals, which yield novel insights for better understanding the molecular basis of human complex disease. We anticipate our approaches will become routine in GWAS downstream analysis and non-coding variant evaluation.


Subject(s)
Deep Learning , Epigenome , Epigenomics/methods , Models, Genetic , Binding Sites , Causality , Chromatin Immunoprecipitation , Datasets as Topic , Genetic Diseases, Inborn/metabolism , Genome-Wide Association Study , Histone Code , Humans , Linkage Disequilibrium , Molecular Sequence Annotation , Organ Specificity , Polymorphism, Single Nucleotide , Transcription Factors/metabolism
11.
J Biol Chem ; 297(4): 101140, 2021 10.
Article in English | MEDLINE | ID: mdl-34461088

ABSTRACT

Biological energy transduction underlies all physiological phenomena in cells. The metabolic systems that support energy transduction have been of great interest due to their association with numerous pathologies including diabetes, cancer, rare genetic diseases, and aberrant cell death. Commercially available bioenergetics technologies (e.g., extracellular flux analysis, high-resolution respirometry, fluorescent dye kits, etc.) have made practical assessment of metabolic parameters widely accessible. This has facilitated an explosion in the number of studies exploring, in particular, the biological implications of oxygen consumption rate (OCR) and substrate level phosphorylation via glycolysis (i.e., via extracellular acidification rate (ECAR)). Though these technologies have demonstrated substantial utility and broad applicability to cell biology research, they are also susceptible to historical assumptions, experimental limitations, and other caveats that have led to premature and/or erroneous interpretations. This review enumerates various important considerations for designing and interpreting cellular and mitochondrial bioenergetics experiments, some common challenges and pitfalls in data interpretation, and some potential "next steps" to be taken that can address these highlighted challenges.


Subject(s)
Diabetes Mellitus/metabolism , Genetic Diseases, Inborn/metabolism , Glycolysis , Mitochondria/metabolism , Models, Biological , Neoplasms/metabolism , Oxidative Phosphorylation , Humans , Oxygen Consumption
12.
Nucleic Acids Res ; 48(20): 11244-11258, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33021629

ABSTRACT

Deletions in mitochondrial DNA (mtDNA) are associated with diverse human pathologies including cancer, aging and mitochondrial disorders. Large-scale deletions span kilobases in length and the loss of these associated genes contributes to crippled oxidative phosphorylation and overall decline in mitochondrial fitness. There is not a united view for how mtDNA deletions are generated and the molecular mechanisms underlying this process are poorly understood. This review discusses the role of replication and repair in mtDNA deletion formation as well as nucleic acid motifs such as repeats, secondary structures, and DNA damage associated with deletion formation in the mitochondrial genome. We propose that while erroneous replication and repair can separately contribute to deletion formation, crosstalk between these pathways is also involved in generating deletions.


Subject(s)
DNA Repair , DNA Replication , DNA, Mitochondrial/biosynthesis , Genetic Diseases, Inborn/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , DNA Breaks, Double-Stranded , DNA Mismatch Repair , DNA, Mitochondrial/metabolism , Genetic Diseases, Inborn/metabolism , Humans , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Oxidative Phosphorylation , Sequence Deletion
13.
Proc Natl Acad Sci U S A ; 116(30): 15236-15243, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31285329

ABSTRACT

Dopamine neurons of the hypothalamic arcuate nucleus (ARC) tonically inhibit the release of the protein hormone prolactin from lactotropic cells in the anterior pituitary gland and thus play a central role in prolactin homeostasis of the body. Prolactin, in turn, orchestrates numerous important biological functions such as maternal behavior, reproduction, and sexual arousal. Here, we identify the canonical transient receptor potential channel Trpc5 as an essential requirement for normal function of dopamine ARC neurons and prolactin homeostasis. By analyzing female mice carrying targeted mutations in the Trpc5 gene including a conditional Trpc5 deletion, we show that Trpc5 is required for maintaining highly stereotyped infraslow membrane potential oscillations of dopamine ARC neurons. Trpc5 is also required for eliciting prolactin-evoked tonic plateau potentials in these neurons that are part of a regulatory feedback circuit. Trpc5 mutant females show severe prolactin deficiency or hypoprolactinemia that is associated with irregular reproductive cyclicity, gonadotropin imbalance, and impaired reproductive capabilities. These results reveal a previously unknown role for the cation channel Trpc5 in prolactin homeostasis of female mice and provide strategies to explore the genetic basis of reproductive disorders and other malfunctions associated with defective prolactin regulation in humans.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Dopaminergic Neurons/metabolism , Genetic Diseases, Inborn/genetics , Lactation Disorders/genetics , Prolactin/deficiency , Prolactin/genetics , TRPC Cation Channels/genetics , Animals , Arcuate Nucleus of Hypothalamus/pathology , Arousal/physiology , Dopaminergic Neurons/pathology , Feedback, Physiological , Female , Gene Expression Regulation , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Gonadotropins/blood , Gonadotropins/genetics , Homeostasis/genetics , Humans , Lactation Disorders/metabolism , Lactation Disorders/pathology , Membrane Potentials/physiology , Mice , Mutation , Prolactin/blood , Prolactin/metabolism , Reproduction/physiology , Signal Transduction , TRPC Cation Channels/deficiency
14.
Int J Mol Sci ; 23(2)2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35054919

ABSTRACT

Inherited retinal diseases (IRDs) are a leading cause of blindness. To date, 260 disease-causing genes have been identified, but there is currently a lack of available and effective treatment options. Cone photoreceptors are responsible for daylight vision but are highly susceptible to disease progression, the loss of cone-mediated vision having the highest impact on the quality of life of IRD patients. Cone degeneration can occur either directly via mutations in cone-specific genes (primary cone death), or indirectly via the primary degeneration of rods followed by subsequent degeneration of cones (secondary cone death). How cones degenerate as a result of pathological mutations remains unclear, hindering the development of effective therapies for IRDs. This review aims to highlight similarities and differences between primary and secondary cone cell death in inherited retinal diseases in order to better define cone death mechanisms and further identify potential treatment options.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Predisposition to Disease , Retinal Cone Photoreceptor Cells/metabolism , Retinal Diseases/genetics , Retinal Diseases/metabolism , Animals , Apoptosis/genetics , Autophagy/genetics , Biomarkers , Cell Death , Endoplasmic Reticulum Stress , Genetic Association Studies , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/therapy , Humans , Oxidative Stress , Retinal Diseases/diagnosis , Retinal Diseases/therapy , Signal Transduction
15.
Curr Opin Hematol ; 28(5): 308-314, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34397590

ABSTRACT

PURPOSE OF REVIEW: Megakaryocytes are rare hematopoietic cells that play an instrumental role in hemostasis, and other important biological processes such as immunity and wound healing. With the advent of cell reprogramming technologies and advances in differentiation protocols, it is now possible to obtain megakaryocytes from any pluripotent stem cell (PSC) via hematopoietic induction. Here, we review recent advances in PSC-derived megakaryocyte (iMK) technology, focusing on platform validation, disease modeling and current limitations. RECENT FINDINGS: A comprehensive study confirmed that iMK can recapitulate many transcriptional and functional aspects of megakaryocyte and platelet biology, including variables associated with complex genetic traits such as sex and race. These findings were corroborated by several pathological models in which iMKs revealed molecular mechanisms behind inherited platelet disorders and assessed the efficacy of novel pharmacological interventions. However, current differentiation protocols generate primarily embryonic iMK, limiting the clinical and translational potential of this system. SUMMARY: iMK are strong candidates to model pathologic mutations involved in platelet defects and develop innovative therapeutic strategies. Future efforts on generating definitive hematopoietic progenitors would improve current platelet generation protocols and expand our capacity to model neonatal and adult megakaryocyte disorders.


Subject(s)
Blood Platelet Disorders , Cell Differentiation , Genetic Diseases, Inborn , Hematopoiesis , Models, Genetic , Pluripotent Stem Cells/metabolism , Animals , Blood Platelet Disorders/genetics , Blood Platelet Disorders/metabolism , Blood Platelet Disorders/therapy , Blood Platelets/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/therapy , Humans , Megakaryocytes/metabolism
16.
EMBO J ; 36(8): 1084-1099, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28264884

ABSTRACT

Properdin (FP) is an essential positive regulator of the complement alternative pathway (AP) providing stabilization of the C3 and C5 convertases, but its oligomeric nature challenges structural analysis. We describe here a novel FP deficiency (E244K) caused by a single point mutation which results in a very low level of AP activity. Recombinant FP E244K is monomeric, fails to support bacteriolysis, and binds weakly to C3 products. We compare this to a monomeric unit excised from oligomeric FP, which is also dysfunctional in bacteriolysis but binds the AP proconvertase, C3 convertase, C3 products and partially stabilizes the convertase. The crystal structure of such a FP-convertase complex suggests that the major contact between FP and the AP convertase is mediated by a single FP thrombospondin repeat and a small region in C3b. Small angle X-ray scattering indicates that FP E244K is trapped in a compact conformation preventing its oligomerization. Our studies demonstrate an essential role of FP oligomerization in vivo while our monomers enable detailed structural insight paving the way for novel modulators of complement.


Subject(s)
Complement C3-C5 Convertases/chemistry , Complement Pathway, Alternative , Properdin/chemistry , Protein Multimerization , Amino Acid Substitution , Complement C3-C5 Convertases/genetics , Complement C3-C5 Convertases/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , Mutation, Missense , Properdin/deficiency , Properdin/genetics , Properdin/metabolism , Protein Domains
17.
J Cell Sci ; 132(20)2019 10 21.
Article in English | MEDLINE | ID: mdl-31636158

ABSTRACT

Adaptor protein (AP) complexes are heterotetramers that select cargo for inclusion into transport vesicles. Five AP complexes (AP-1 to AP-5) have been described, each with a distinct localisation and function. Furthermore, patients with a range of disorders, particularly involving the nervous system, have now been identified with mutations in each of the AP complexes. In many cases this has been correlated with aberrantly localised membrane proteins. In this Cell Science at a Glance article and the accompanying poster, we summarize what is known about the five AP complexes and discuss how this helps to explain the clinical features of the different genetic disorders.


Subject(s)
Adaptor Proteins, Vesicular Transport , Genetic Diseases, Inborn , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans
18.
Blood ; 134(23): 2070-2081, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31217188

ABSTRACT

To identify novel causes of hereditary thrombocytopenia, we performed a genetic association analysis of whole-genome sequencing data from 13 037 individuals enrolled in the National Institute for Health Research (NIHR) BioResource, including 233 cases with isolated thrombocytopenia. We found an association between rare variants in the transcription factor-encoding gene IKZF5 and thrombocytopenia. We report 5 causal missense variants in or near IKZF5 zinc fingers, of which 2 occurred de novo and 3 co-segregated in 3 pedigrees. A canonical DNA-zinc finger binding model predicts that 3 of the variants alter DNA recognition. Expression studies showed that chromatin binding was disrupted in mutant compared with wild-type IKZF5, and electron microscopy revealed a reduced quantity of α granules in normally sized platelets. Proplatelet formation was reduced in megakaryocytes from 7 cases relative to 6 controls. Comparison of RNA-sequencing data from platelets, monocytes, neutrophils, and CD4+ T cells from 3 cases and 14 healthy controls showed 1194 differentially expressed genes in platelets but only 4 differentially expressed genes in each of the other blood cell types. In conclusion, IKZF5 is a novel transcriptional regulator of megakaryopoiesis and the eighth transcription factor associated with dominant thrombocytopenia in humans.


Subject(s)
Blood Platelets , Genetic Diseases, Inborn , Germ-Line Mutation , Ikaros Transcription Factor , Mutation, Missense , Thrombocytopenia , Thrombopoiesis/genetics , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Chromatin/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Female , Gene Expression Regulation , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , HEK293 Cells , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Male , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
19.
Am J Med Genet A ; 185(8): 2576-2581, 2021 08.
Article in English | MEDLINE | ID: mdl-33973706

ABSTRACT

Hypoxia-inducible factors (HIFs) activate gene transcription in response to reduced O2 availability and play critical roles in development, physiology, and disease pathogenesis. Mutations that dysregulate HIF activity are the genetic basis for tumor predisposition in the von Hippel-Lindau syndrome and excess red blood cell production in hereditary erythrocytosis.


Subject(s)
Genetic Association Studies , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Predisposition to Disease , Oxygen/metabolism , Phenotype , Biomarkers , Diagnosis, Differential , Genetic Diseases, Inborn/diagnosis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Polycythemia/congenital , Polycythemia/diagnosis , Polycythemia/metabolism , Signal Transduction , von Hippel-Lindau Disease/diagnosis , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/metabolism
20.
Am J Med Genet A ; 185(11): 3334-3339, 2021 11.
Article in English | MEDLINE | ID: mdl-34655169

ABSTRACT

Hypoxia-inducible factors (HIFs) activate gene transcription in response to reduced O2 availability and play critical roles in development, physiology, and disease pathogenesis. Mutations that dysregulate HIF activity are the genetic basis for tumor predisposition in the von Hippel-Lindau syndrome and excess red blood cell production in hereditary erythrocytosis.


Subject(s)
Genetic Diseases, Inborn/genetics , Oxygen/metabolism , Polycythemia/congenital , von Hippel-Lindau Disease/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Genetic Diseases, Inborn/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mutation/genetics , Polycythemia/genetics , Polycythemia/metabolism , Polycythemia/pathology , von Hippel-Lindau Disease/metabolism , von Hippel-Lindau Disease/pathology
SELECTION OF CITATIONS
SEARCH DETAIL