Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 625
Filter
1.
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
2.
Orphanet J Rare Dis ; 18(1): 130, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37259144

ABSTRACT

Pulmonary alveolar microlithiasis (PAM) is a rare autosomal recessive lung disease caused by variants in the SLC34A2 gene encoding the sodium-dependent phosphate transport protein 2B, NaPi-2b. PAM is characterized by deposition of calcium phosphate crystals in the alveoli. Onset and clinical course vary considerably; some patients remain asymptomatic while others develop severe respiratory failure with a significant symptom burden and compromised survival. It is likely that PAM is under-reported due to lack of recognition, misdiagnosis, and mild clinical presentation. Most patients are genetically uncharacterized as the diagnostic confirmation of PAM has traditionally not included a genetic analysis. Genetic testing may in the future be the preferred tool for diagnostics instead of invasive methods. This systematic review aims to provide an overview of the growing knowledge of PAM genetics. Rare variants in SLC34A2 are found in almost all genetically tested patients. So far, 34 allelic variants have been identified in at least 68 patients. A majority of these are present in the homozygous state; however, a few are found in the compound heterozygous form. Most of the allelic variants involve only a single nucleotide. Half of the variants are either nonsense or frameshifts, resulting in premature termination of the protein or decay of the mRNA. There is currently no cure for PAM, and the only effective treatment is lung transplantation. Management is mainly symptomatic, but an improved understanding of the underlying pathophysiology will hopefully result in development of targeted treatment options. More standardized data on PAM patients, including a genetic diagnosis covering larger international populations, would support the design and implementation of clinical studies to the benefit of patients. Further genetic characterization and understanding of how the molecular changes influence disease phenotype will hopefully allow earlier diagnosis and treatment of the disease in the future.


Subject(s)
Calcinosis , Genetic Diseases, Inborn , Lung Diseases , Humans , Lung Diseases/genetics , Lung , Calcinosis/genetics , Frameshift Mutation , Pulmonary Alveoli/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIb/metabolism
3.
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
4.
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
5.
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
6.
Int J Mol Sci ; 22(24)2021 Dec 12.
Article in English | MEDLINE | ID: mdl-34948153

ABSTRACT

Rare genetic diseases are a group of pathologies with often unmet clinical needs. Even if rare by a single genetic disease (from 1/2000 to 1/more than 1,000,000), the total number of patients concerned account for approximatively 400 million peoples worldwide. Finding treatments remains challenging due to the complexity of these diseases, the small number of patients and the challenge in conducting clinical trials. Therefore, innovative preclinical research strategies are required. The zebrafish has emerged as a powerful animal model for investigating rare diseases. Zebrafish combines conserved vertebrate characteristics with high rate of breeding, limited housing requirements and low costs. More than 84% of human genes responsible for diseases present an orthologue, suggesting that the majority of genetic diseases could be modelized in zebrafish. In this review, we emphasize the unique advantages of zebrafish models over other in vivo models, particularly underlining the high throughput phenotypic capacity for therapeutic screening. We briefly introduce how the generation of zebrafish transgenic lines by gene-modulating technologies can be used to model rare genetic diseases. Then, we describe how zebrafish could be phenotyped using state-of-the-art technologies. Two prototypic examples of rare diseases illustrate how zebrafish models could play a critical role in deciphering the underlying mechanisms of rare genetic diseases and their use to identify innovative therapeutic solutions.


Subject(s)
Genetic Diseases, Inborn , Models, Genetic , Rare Diseases , Zebrafish , Animals , Biomedical Research , Disease Models, Animal , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/therapy , Humans , Rare Diseases/genetics , Rare Diseases/metabolism , Rare Diseases/therapy , Zebrafish/genetics , Zebrafish/metabolism
7.
Front Immunol ; 12: 717388, 2021.
Article in English | MEDLINE | ID: mdl-34603291

ABSTRACT

Among inborn errors of immunity (IEIs), some conditions are characterized by inflammation and autoimmunity at the front line and are particularly challenging to treat. Monogenic diseases associated with gain-of-function mutations in genes critical for cytokine signaling through the JAK-STAT pathway belong to this group. These conditions represent good candidates for treatment with JAK inhibitors. Type I interferonopathies, a group of recently identified monogenic auto-inflammatory diseases characterized by excessive secretion of type I IFN, are also good candidates with growing experiences reported in the literature. However, many questions remain regarding the choice of the drug, the dose (in particular in children), the efficacy on the various manifestations, the monitoring of the treatment, and the management of potent side effects in particular in patients with infectious susceptibility. This review will summarize the current experiences reported and will highlight the unmet needs.


Subject(s)
Disease Management , Disease Susceptibility , Genetic Diseases, Inborn/drug therapy , Immune System Diseases/drug therapy , Janus Kinase Inhibitors/therapeutic use , Molecular Targeted Therapy , Animals , Biomarkers , Clinical Studies as Topic , Drug Development , Drug Evaluation, Preclinical , Genetic Diseases, Inborn/etiology , Genetic Diseases, Inborn/metabolism , Humans , Immune System Diseases/etiology , Immune System Diseases/metabolism , Janus Kinase Inhibitors/administration & dosage , Janus Kinase Inhibitors/adverse effects , Janus Kinases/genetics , Janus Kinases/metabolism , Mutation , STAT Transcription Factors/metabolism , Signal Transduction
8.
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
9.
Int J Mol Sci ; 22(17)2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34502077

ABSTRACT

The cloning of connexins cDNA opened the way to the field of gap junction channelopathies. Thus far, at least 35 genetic diseases, resulting from mutations of 11 different connexin genes, are known to cause numerous structural and functional defects in the central and peripheral nervous system as well as in the heart, skin, eyes, teeth, ears, bone, hair, nails and lymphatic system. While all of these diseases are due to connexin mutations, minimal attention has been paid to the potential diseases of cell-cell communication caused by mutations of Cx-associated molecules. An important Cx accessory protein is calmodulin (CaM), which is the major regulator of gap junction channel gating and a molecule relevant to gap junction formation. Recently, diseases caused by CaM mutations (calmodulinopathies) have been identified, but thus far calmodulinopathy studies have not considered the potential effect of CaM mutations on gap junction function. The major goal of this review is to raise awareness on the likely role of CaM mutations in defects of gap junction mediated cell communication. Our studies have demonstrated that certain CaM mutants affect gap junction channel gating or expression, so it would not be surprising to learn that CaM mutations known to cause diseases also affect cell communication mediated by gap junction channels.


Subject(s)
Calmodulin/genetics , Channelopathies/genetics , Connexins/genetics , Genetic Diseases, Inborn/genetics , Animals , Calmodulin/metabolism , Cell Communication , Channelopathies/metabolism , Connexins/metabolism , Genetic Diseases, Inborn/metabolism , Humans , Mutation
10.
Clin Biochem ; 98: 78-80, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34529996

ABSTRACT

This report investigates an unusual case of recurrent pancreatitis. A 22-year-old female was admitted to the emergency room for severe abdominal pain, nausea, and weight loss. She reported having these symptoms since she was a toddler. The clinician ordered fecal pancreatic elastase-1, fat-soluble vitamins, molecular studies, and imaging of the pancreas by computed tomography. The screening test result for fecal pancreatic elastase-1 revealed severe pancreatic exocrine insufficiency, and the concentrations of fat-soluble vitamins were also low. Imaging showed scattered calcifications in the pancreas. These findings supported a diagnosis of chronic pancreatitis. Due to the rarity of chronic pancreatitis in young adults, molecular studies were performed. The patient was found to be homozygous for a mutation in the SPINK1 gene, which is associated with hereditary pancreatitis. This case report discusses hereditary pancreatitis and highlights data on the utilization of fecal pancreatic elastase-1 to assess pancreatic exocrine insufficiency.


Subject(s)
Genetic Diseases, Inborn/genetics , Homozygote , Mutation , Pancreatitis, Chronic/genetics , Trypsin Inhibitor, Kazal Pancreatic/genetics , Adult , Female , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/metabolism , Humans , Pancreatic Elastase/genetics , Pancreatic Elastase/metabolism , Pancreatitis, Chronic/diagnosis , Pancreatitis, Chronic/metabolism , Trypsin Inhibitor, Kazal Pancreatic/metabolism , Young Adult
11.
Front Immunol ; 12: 680334, 2021.
Article in English | MEDLINE | ID: mdl-34421895

ABSTRACT

Background: Inborn errors of immunity (IEI) present with a large phenotypic spectrum of disease, which can pose diagnostic and therapeutic challenges. Suppressor of cytokine signaling 1 (SOCS1) is a key negative regulator of cytokine signaling, and has recently been associated with a novel IEI. Of patients described to date, it is apparent that SOCS1 haploinsufficiency has a pleiotropic effect in humans. Objective: We sought to investigate whether dysregulation of immune pathways, in addition to STAT1, play a role in the broad clinical manifestations of SOCS1 haploinsufficiency. Methods: We assessed impacts of reduced SOCS1 expression across multiple immune cell pathways utilizing patient cells and CRISPR/Cas9 edited primary human T cells. Results: SOCS1 haploinsufficiency phenotypes straddled across the International Union of Immunological Societies classifications of IEI. We found that reduced SOCS1 expression led to dysregulation of multiple intracellular pathways in immune cells. STAT1 phosphorylation is enhanced, comparably with STAT1 gain-of-function mutations, and STAT3 phosphorylation is similarly reduced with concurrent reduction of Th17 cells. Furthermore, reduced SOCS1 E3 ligase function was associated with increased FAK1 in immune cells, and increased AKT and p70 ribosomal protein S6 kinase phosphorylation. We also found Toll-like receptor responses are increased in SOCS1 haploinsufficiency patients. Conclusions: SOCS1 haploinsufficiency is a pleiotropic monogenic IEI. Dysregulation of multiple immune cell pathways may explain the variable clinical phenotype associated with this new condition. Knowledge of these additional dysregulated immune pathways is important when considering the optimum management for SOCS1 haploinsufficient patients.


Subject(s)
Haploinsufficiency , Immune System/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Alleles , Autoimmunity , Biomarkers , Case-Control Studies , Child , Child, Preschool , Cytokines , Female , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , Job Syndrome/diagnosis , Job Syndrome/etiology , Job Syndrome/metabolism , Male , Models, Biological , Pedigree , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
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
13.
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
14.
Int J Mol Sci ; 22(13)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203525

ABSTRACT

eEF1A1 and eEF1A2 are paralogous proteins whose presence in most normal eukaryotic cells is mutually exclusive and developmentally regulated. Often described in the scientific literature under the collective name eEF1A, which stands for eukaryotic elongation factor 1A, their best known activity (in a monomeric, GTP-bound conformation) is to bind aminoacyl-tRNAs and deliver them to the A-site of the 80S ribosome. However, both eEF1A1 and eEF1A2 are endowed with multitasking abilities (sometimes performed by homo- and heterodimers) and can be located in different subcellular compartments, from the plasma membrane to the nucleus. Given the high sequence identity of these two sister proteins and the large number of post-translational modifications they can undergo, we are often confronted with the dilemma of discerning which is the particular proteoform that is actually responsible for the ascribed biochemical or cellular effects. We argue in this review that acquiring this knowledge is essential to help clarify, in molecular and structural terms, the mechanistic involvement of these two ancestral and abundant G proteins in a variety of fundamental cellular processes other than translation elongation. Of particular importance for this special issue is the fact that several de novo heterozygous missense mutations in the human EEF1A2 gene are associated with a subset of rare but severe neurological syndromes and cardiomyopathies.


Subject(s)
GTP-Binding Proteins/metabolism , Peptide Elongation Factor 1/metabolism , Animals , GTP-Binding Proteins/genetics , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , Mutation/genetics , Peptide Elongation Factor 1/genetics , Protein Binding , Protein Processing, Post-Translational
15.
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
16.
Theranostics ; 11(15): 7262-7275, 2021.
Article in English | MEDLINE | ID: mdl-34158849

ABSTRACT

Rationale: Congenital biliary atresia (BA) is a destructive obliterative cholangiopathy of neonates that affects both intrahepatic and extrahepatic bile ducts. However, the cause of BA is largely unknown. Methods: We explored the cell junctions and polarity complexes in early biopsy BA livers by immunofluorescence staining and western blot. Cdc42, as a key cell junction and polarity regulator, was found dramatically decreased in BA livers. Therefore, in order to investigate the role of Cdc42 in BA development, we constructed liver-specific and tamoxifen induced cholangiocyte-specific Cdc42 deleted transgenic mice. We further evaluated the role of bile acid in aggravating biliary damage in Cdc42 insufficient mouse liver. Results: We found a dramatic defect in the assembly of cell junctions and polarity complexes in both cholangiocytes and hepatocytes in BA livers. This defect was characterized by the disordered location of cell junction proteins, including ZO1, ß-catenin, E-cadherin and claudin-3. Cdc42 and its active form, Cdc42-GTP, which serves as a small Rho GTPase to orchestrate the assembly of polarity complexes with Par6/Par3/αPKC, were substantially reduced in BA livers. Selective Cdc42 deficiency in fetal mouse cholangiocytes resulted in histological changes similar to those found in human BA livers, including obstruction in both the intra- and extrahepatic bile ducts, epithelial atrophy, and the disruption of cell junction and polarity complexes. A reduction in bile acids notably improved the histology and serological indices in Cdc42-mutant mice. Conclusion: Our results illustrate that BA is closely correlated with the impaired assembly of cell junction and polarity complexes in liver cells, which is likely caused by Cdc42 insufficiency and aggravated by bile acid corrosion.


Subject(s)
Biliary Atresia , Genetic Diseases, Inborn , Intercellular Junctions , Liver/metabolism , cdc42 GTP-Binding Protein/deficiency , Animals , Biliary Atresia/genetics , Biliary Atresia/metabolism , Biliary Atresia/pathology , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Humans , Infant , Intercellular Junctions/genetics , Intercellular Junctions/metabolism , Intercellular Junctions/pathology , Liver/pathology , Male , Mice , Mice, Knockout , cdc42 GTP-Binding Protein/metabolism
17.
Sci Rep ; 11(1): 11432, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34075074

ABSTRACT

Retinitis Pigmentosa (RP) is a mostly incurable inherited retinal degeneration affecting approximately 1 in 4000 individuals globally. The goal of this work was to identify drugs that can help patients suffering from the disease. To accomplish this, we screened drugs on a zebrafish autosomal dominant RP model. This model expresses a truncated human rhodopsin transgene (Q344X) causing significant rod degeneration by 7 days post-fertilization (dpf). Consequently, the larvae displayed a deficit in visual motor response (VMR) under scotopic condition. The diminished VMR was leveraged to screen an ENZO SCREEN-WELL REDOX library since oxidative stress is postulated to play a role in RP progression. Our screening identified a beta-blocker, carvedilol, that ameliorated the deficient VMR of the RP larvae and increased their rod number. Carvedilol may directly on rods as it affected the adrenergic pathway in the photoreceptor-like human Y79 cell line. Since carvedilol is an FDA-approved drug, our findings suggest that carvedilol can potentially be repurposed to treat autosomal dominant RP patients.


Subject(s)
Animals, Genetically Modified , Behavior, Animal/drug effects , Genetic Diseases, Inborn , Retinitis Pigmentosa , Rhodopsin , Vision, Ocular , Zebrafish , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Cell Line , Drug Evaluation, Preclinical , Genetic Diseases, Inborn/drug therapy , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , Mutation , Retinal Rod Photoreceptor Cells , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Transgenes , Vision, Ocular/drug effects , Vision, Ocular/immunology , Zebrafish/genetics , Zebrafish/metabolism
18.
Pathol Int ; 71(8): 521-529, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34166554

ABSTRACT

The Notch signaling pathway plays a key role in the morphogenesis of the biliary tree, but its involvement in cystic biliary diseases, such as Caroli disease (CD) and polycystic liver disease (PLD), has yet to be determined. Immunostaining was performed using liver sections of CD and PLD, and the results were compared with those of congenital hepatic fibrosis (CHF) and von Meyenburg complex (VMC). The expression of Notch receptor 1 (Notch1) was increased in the nuclei of biliary epithelial cells in all cases of CD and PLD, whereas it remained at a low level in CHF and VMC. In addition, Notch2 and Notch3 were preferably expressed in the nuclei of biliary epithelial cells of PLD. Accordingly, the Notch effector Hes1 was highly expressed in biliary epithelial cells of CD and PLD, and the cell proliferative activity was significantly higher in CD and PLD. The expression of the Notch ligand Delta-like 1 was significantly increased in biliary epithelial cells of CD and PLD, which may be causally associated with the nuclear overexpression of Notch1 and Hes1. These results indicate that aberrant activation of the Notch-Hes1 signaling pathway may be responsible for the progression of biliary cystogenesis in CD and PLD.


Subject(s)
Caroli Disease , Cysts , Liver Diseases , Receptor, Notch2/metabolism , Caroli Disease/metabolism , Caroli Disease/pathology , Cysts/metabolism , Cysts/pathology , Epithelial Cells/metabolism , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Humans , Immunohistochemistry , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Signal Transduction , Transcription Factor HES-1/metabolism
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.
Nat Commun ; 12(1): 2344, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879794

ABSTRACT

Direct determination of RNA structures and interactions in living cells is critical for understanding their functions in normal physiology and disease states. Here, we present PARIS2, a dramatically improved method for RNA duplex determination in vivo with >4000-fold higher efficiency than previous methods. PARIS2 captures ribosome binding sites on mRNAs, reporting translation status on a transcriptome scale. Applying PARIS2 to the U8 snoRNA mutated in the neurological disorder LCC, we discover a network of dynamic RNA structures and interactions which are destabilized by patient mutations. We report the first whole genome structure of enterovirus D68, an RNA virus that causes polio-like symptoms, revealing highly dynamic conformations altered by antiviral drugs and different pathogenic strains. We also discover a replication-associated asymmetry on the (+) and (-) strands of the viral genome. This study establishes a powerful technology for efficient interrogation of the RNA structurome and interactome in human diseases.


Subject(s)
Communicable Diseases/genetics , Communicable Diseases/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Photochemistry/methods , RNA/chemistry , RNA/metabolism , Calcinosis/genetics , Calcinosis/metabolism , Central Nervous System Cysts/genetics , Central Nervous System Cysts/metabolism , Cross-Linking Reagents , Enterovirus D, Human/genetics , Furocoumarins , Genome, Viral , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Models, Molecular , Mutation , Nucleic Acid Conformation , Photochemical Processes , RNA/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...