Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters











Publication year range
1.
Stem Cell Res Ther ; 14(1): 345, 2023 12 05.
Article in English | MEDLINE | ID: mdl-38049901

ABSTRACT

BACKGROUND: Genomic sequencing in congenital heart disease (CHD) patients often discovers novel genetic variants, which are classified as variants of uncertain significance (VUS). Functional analysis of each VUS is required in specialised laboratories, to determine whether the VUS is disease causative or not, leading to lengthy diagnostic delays. We investigated stem cell cardiac disease modelling and transcriptomics for the purpose of genetic variant classification using a GATA4 (p.Arg283Cys) VUS in a patient with CHD. METHODS: We performed high efficiency CRISPR gene editing with homology directed repair in induced pluripotent stem cells (iPSCs), followed by rapid clonal selection with amplicon sequencing. Genetic variant and healthy matched control cells were compared using cardiomyocyte disease modelling and transcriptomics. RESULTS: Genetic variant and healthy cardiomyocytes similarly expressed Troponin T (cTNNT), and GATA4. Transcriptomics analysis of cardiomyocyte differentiation identified changes consistent with the patient's clinical human phenotype ontology terms. Further, transcriptomics revealed changes in calcium signalling, and cardiomyocyte adrenergic signalling in the variant cells. Functional testing demonstrated, altered action potentials in GATA4 genetic variant cardiomyocytes were consistent with patient cardiac abnormalities. CONCLUSIONS: This work provides in vivo functional studies supportive of a damaging effect on the gene or gene product. Furthermore, we demonstrate the utility of iPSCs, CRISPR gene editing and cardiac disease modelling for genetic variant interpretation. The method can readily be applied to other genetic variants in GATA4 or other genes in cardiac disease, providing a centralised assessment pathway for patient genetic variant interpretation.


Subject(s)
Gene Editing , Heart Defects, Congenital , Humans , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Myocytes, Cardiac/metabolism , Base Sequence , Signal Transduction
2.
Sci Rep ; 13(1): 11296, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37438479

ABSTRACT

Familial hypertrophic cardiomyopathy (FHC) patients are advised to avoid strenuous exercise due to increased risk of arrhythmias. Mice expressing the human FHC-causing mutation R403Q in the myosin heavy chain gene (MYH6) recapitulate the human phenotype, including cytoskeletal disarray and increased arrhythmia susceptibility. Following in vivo administration of isoproterenol, mutant mice exhibited tachyarrhythmias, poor recovery and fatigue. Arrhythmias were attenuated with the ß-blocker atenolol and protein kinase A inhibitor PKI. Mutant cardiac myocytes had significantly prolonged action potentials and triggered automaticity due to reduced repolarization reserve and connexin 43 expression. Isoproterenol shortened cycle length, and escalated electrical instability. Surprisingly isoproterenol did not increase CaV1.2 current. We found alterations in CaV1.2-ß1 adrenergic receptor colocalization assessed using super-resolution nanoscopy, and increased CaV1.2 phosphorylation in mutant hearts. Our results reveal for the first time that altered ion channel expression, co-localization and ß-adrenergic receptor signaling associated with myocyte disarray contribute to electrical instability in the R403Q mutant heart.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial , Cardiomyopathy, Hypertrophic , Humans , Animals , Mice , Isoproterenol , Cardiomyopathy, Hypertrophic/genetics , Arrhythmias, Cardiac , Heart
3.
Commun Biol ; 6(1): 4, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36596888

ABSTRACT

Hypertrophic cardiomyopathy is an inherited disorder due to mutations in contractile proteins that results in a stiff, hypercontractile myocardium. To understand the role of cardiac stiffness in disease progression, here we create an in vitro model of hypertrophic cardiomyopathy utilizing hydrogel technology. Culturing wild-type cardiac myocytes on hydrogels with a Young's Moduli (stiffness) mimicking hypertrophic cardiomyopathy myocardium is sufficient to induce a hypermetabolic mitochondrial state versus myocytes plated on hydrogels simulating healthy myocardium. Significantly, these data mirror that of myocytes isolated from a murine model of human hypertrophic cardiomyopathy (cTnI-G203S). Conversely, cTnI-G203S myocyte mitochondrial function is completely restored when plated on hydrogels mimicking healthy myocardium. We identify a mechanosensing feedback mechanism between the extracellular matrix and cytoskeletal network that regulates mitochondrial function under healthy conditions, but participates in the progression of hypertrophic cardiomyopathy pathophysiology resulting from sarcomeric gene mutations. Importantly, we pinpoint key 'linker' sites in this schema that may represent potential therapeutic targets.


Subject(s)
Cardiomyopathy, Hypertrophic , Mice , Humans , Animals , Feedback , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cytoskeleton/metabolism , Myocytes, Cardiac/metabolism , Troponin I/genetics , Troponin I/metabolism , Extracellular Matrix/metabolism , Hydrogels
4.
Sci Adv ; 7(39): eabi7514, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34559558

ABSTRACT

Mitochondrial energy metabolism plays an important role in the pathophysiology of insulin resistance. Recently, a missense N437S variant was identified in the MRPP3 gene, which encodes a mitochondrial RNA processing enzyme within the RNase P complex, with predicted impact on metabolism. We used CRISPR-Cas9 genome editing to introduce this variant into the mouse Mrpp3 gene and show that the variant causes insulin resistance on a high-fat diet. The variant did not influence mitochondrial gene expression markedly, but instead, it reduced mitochondrial calcium that lowered insulin release from the pancreatic islet ß cells of the Mrpp3 variant mice. Reduced insulin secretion resulted in lower insulin levels that contributed to imbalanced metabolism and liver steatosis in the Mrpp3 variant mice on a high-fat diet. Our findings reveal that the MRPP3 variant may be a predisposing factor to insulin resistance and metabolic disease in the human population.

5.
Aging Cell ; 20(7): e13408, 2021 07.
Article in English | MEDLINE | ID: mdl-34096683

ABSTRACT

Changes in the rate and fidelity of mitochondrial protein synthesis impact the metabolic and physiological roles of mitochondria. Here we explored how environmental stress in the form of a high-fat diet modulates mitochondrial translation and affects lifespan in mutant mice with error-prone (Mrps12ep/ep ) or hyper-accurate (Mrps12ha/ha ) mitochondrial ribosomes. Intriguingly, although both mutations are metabolically beneficial in reducing body weight, decreasing circulating insulin and increasing glucose tolerance during a high-fat diet, they manifest divergent (either deleterious or beneficial) outcomes in a tissue-specific manner. In two distinct organs that are commonly affected by the metabolic disease, the heart and the liver, Mrps12ep/ep mice were protected against heart defects but sensitive towards lipid accumulation in the liver, activating genes involved in steroid and amino acid metabolism. In contrast, enhanced translational accuracy in Mrps12ha/ha mice protected the liver from a high-fat diet through activation of liver proliferation programs, but enhanced the development of severe hypertrophic cardiomyopathy and led to reduced lifespan. These findings reflect the complex transcriptional and cell signalling responses that differ between post-mitotic (heart) and highly proliferative (liver) tissues. We show trade-offs between the rate and fidelity of mitochondrial protein synthesis dictate tissue-specific outcomes due to commonly encountered stressful environmental conditions or aging.


Subject(s)
Cardiovascular Diseases/genetics , Mitochondria/metabolism , Stress, Physiological/genetics , Animals , Humans , Longevity , Male , Mice
6.
J Physiol ; 599(14): 3495-3512, 2021 07.
Article in English | MEDLINE | ID: mdl-32822065

ABSTRACT

Sarcomeric gene mutations are associated with the development of hypertrophic cardiomyopathy (HCM). Current drug therapeutics for HCM patients are effective in relieving symptoms, but do not prevent or reverse disease progression. Moreover, due to heterogeneity in the clinical manifestations of the disease, patients experience variable outcomes in response to therapeutics. Mechanistically, alterations in calcium handling, sarcomeric disorganization, energy metabolism and contractility participate in HCM disease progression. While some similarities exist, each mutation appears to lead to mutation-specific pathophysiology. Furthermore, these alterations may precede or proceed development of the pathology. This review assesses the efficacy of HCM therapeutics from studies performed in animal models of HCM and human clinical trials. Evidence suggests that a preventative rather than corrective therapeutic approach may be more efficacious in the treatment of HCM. In addition, a clear understanding of mutation-specific mechanisms may assist in informing the most effective therapeutic mode of action.


Subject(s)
Cardiomyopathy, Hypertrophic , Animals , Calcium/metabolism , Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/genetics , Energy Metabolism , Humans , Mutation , Sarcomeres/metabolism
7.
J Physiol ; 599(14): 3449-3462, 2021 07.
Article in English | MEDLINE | ID: mdl-32710561

ABSTRACT

The evolutionary acquisition of mitochondria has given rise to the diversity of eukaryotic life. Mitochondria have retained their ancestral α-proteobacterial traits through the maintenance of double membranes and their own circular genome. Their genome varies in size from very large in plants to the smallest in animals and their parasites. The mitochondrial genome encodes essential genes for protein synthesis and has to coordinate its expression with the nuclear genome from which it sources most of the proteins required for mitochondrial biogenesis and function. The mitochondrial protein synthesis machinery is unique because it is encoded by both the nuclear and mitochondrial genomes thereby requiring tight regulation to produce the respiratory complexes that drive oxidative phosphorylation for energy production. The fidelity and coordination of mitochondrial protein synthesis are essential for ATP production. Here we compare and contrast the mitochondrial translation mechanisms in mammals and fungi to bacteria and reveal that their diverse regulation can have unusual impacts on the health and disease of these organisms. We highlight that in mammals the rate of protein synthesis is more important than the fidelity of translation, enabling coordinated biogenesis of the mitochondrial respiratory chain with respiratory chain proteins synthesised by cytoplasmic ribosomes. Changes in mitochondrial protein fidelity can trigger the activation of the diverse cellular signalling networks in fungi and mammals to combat dysfunction in energy conservation. The physiological consequences of altered fidelity of protein synthesis can range from liver regeneration to the onset and development of cardiomyopathy.


Subject(s)
Genome, Mitochondrial , Protein Biosynthesis , Animals , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Ribosomes/metabolism
8.
Proc Natl Acad Sci U S A ; 117(37): 23113-23124, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32859761

ABSTRACT

Currently there is an unmet need for treatments that can prevent hypertrophic cardiomyopathy (HCM). Using a murine model we previously identified that HCM causing cardiac troponin I mutation Gly203Ser (cTnI-G203S) is associated with increased mitochondrial metabolic activity, consistent with the human condition. These alterations precede development of the cardiomyopathy. Here we examine the efficacy of in vivo treatment of cTnI-G203S mice with a peptide derived against the α-interaction domain of the cardiac L-type calcium channel (AID-TAT) on restoring mitochondrial metabolic activity, and preventing HCM. cTnI-G203S or age-matched wt mice were treated with active or inactive AID-TAT. Following treatment, targeted metabolomics was utilized to evaluate myocardial substrate metabolism. Cardiac myocyte mitochondrial metabolic activity was assessed as alterations in mitochondrial membrane potential and flavoprotein oxidation. Cardiac morphology and function were examined using echocardiography. Cardiac uptake was assessed using an in vivo multispectral imaging system. We identified alterations in six biochemical intermediates in cTnI-G203S hearts consistent with increased anaplerosis. We also reveal that AID-TAT treatment of precardiomyopathic cTnI-G203S mice, but not mice with established cardiomyopathy, restored cardiac myocyte mitochondrial membrane potential and flavoprotein oxidation, and prevented myocardial hypertrophy. Importantly, AID-TAT was rapidly targeted to the heart, and not retained by the liver or kidneys. Overall, we identify biomarkers of HCM resulting from the cTnI mutation Gly203Ser, and present a safe, preventative therapy for associated cardiomyopathy. Utilizing AID-TAT to modulate cardiac metabolic activity may be beneficial in preventing HCM in "at risk" patients with identified Gly203Ser gene mutations.


Subject(s)
Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Disease Models, Animal , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation/genetics , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Peptides/pharmacology , Troponin I/metabolism
9.
Heart Lung Circ ; 29(11): 1588-1595, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32839116

ABSTRACT

BACKGROUND: Cardiovascular disease is the leading cause of death in Australia. Investment in research solutions has been demonstrated to yield health and a 9.8-fold return economic benefit. The sector, however, is severely challenged with success rates of traditional peer-reviewed funding in decline. Here, we aimed to understand the perceived challenges faced by the cardiovascular workforce in Australia prior to the COVID-19 pandemic. METHODS: We used an online survey distributed across Australian cardiovascular societies/councils, universities and research institutes over a period of 6 months during 2019, with 548 completed responses. Inclusion criteria included being an Australian resident or an Australian citizen who lived overseas, and a current or past student or employee in the field of cardiovascular research. RESULTS: The mean age of respondents was 42±13 years, 47% were male, 85% had a full-time position, and 40% were a group leader or laboratory head. Twenty-three per cent (23%) had permanent employment, and 82% of full-time workers regularly worked >40 hours/week. Sixty-eight per cent (68%) said they had previously considered leaving the cardiovascular research sector. If their position could not be funded in the next few years, a staggering 91% of respondents would leave the sector. Compared to PhD- and age-matched men, women were less likely to be a laboratory head and to feel they had a long-term career path as a cardiovascular researcher, while more women were unsure about future employment and had considered leaving the sector (all p<0.05). Greater job security (76%) and government and philanthropic investment in cardiovascular research (72%) were highlighted by responders as the main changes to current practices that would encourage them to stay. CONCLUSION: Strategic solutions, such as diversification of career pathways and funding sources, and moving from a competitive to a collaborative culture, need to be a priority to decrease reliance on government funding and allow cardiovascular researchers to thrive.


Subject(s)
Biomedical Research , Cardiovascular Diseases , Coronavirus Infections/epidemiology , Financial Management , Pneumonia, Viral/epidemiology , Research Personnel , Research Support as Topic , Workforce , Adult , Australia , Betacoronavirus , Biomedical Research/economics , Biomedical Research/organization & administration , Biomedical Research/trends , COVID-19 , Employment/economics , Employment/psychology , Female , Financial Management/methods , Financial Management/organization & administration , Financial Management/statistics & numerical data , Financing, Government , Humans , Male , Organizational Culture , Pandemics , Planning Techniques , Research Personnel/economics , Research Personnel/psychology , Research Personnel/statistics & numerical data , Research Support as Topic/organization & administration , Research Support as Topic/trends , SARS-CoV-2 , Surveys and Questionnaires , Workforce/statistics & numerical data
10.
Nanomedicine ; 29: 102264, 2020 10.
Article in English | MEDLINE | ID: mdl-32659322

ABSTRACT

Therapeutic approaches for myocardial ischemia-reperfusion injury (MI) have been ineffective due to limited bioavailability and poor specificity. We have previously shown that a peptide that targets the α-interaction domain of the cardiac L-type calcium channel (AID-peptide) attenuates MI when tethered to transactivator of transcription sequence (TAT) or spherical nanoparticles. However some reservations remain regarding use of these delivery platforms due to the relationship with human immunodeficiency virus, off-target effects and toxicity. Here we investigate the use of linear dendronized polymers (denpols) to deliver AID-peptide as a potential MI therapy using in vitro, ex vivo and in vivo models. Optimized denpol-complexed AID-peptide facilitated in vitro cardiac uptake of AID-peptide, and reduced MI. Maximal in vivo cardiac uptake was achieved within the 2 h therapeutic time window for acute myocardial infarction. Importantly, optimized denpol-complexed AID-peptide was not toxic. This platform may represent an alternative therapeutic approach for the prevention of MI.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Heart/drug effects , Myocardial Reperfusion Injury/drug therapy , Nanoparticles/chemistry , Animals , Calcium Channel Blockers/chemistry , Calcium Channels, L-Type/drug effects , Disease Models, Animal , Guinea Pigs , Heart/physiopathology , Humans , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Peptides/chemistry , Peptides/pharmacology , Polymers/chemistry , Polymers/pharmacology
11.
FEBS Lett ; 593(22): 3149-3161, 2019 11.
Article in English | MEDLINE | ID: mdl-31677274

ABSTRACT

Scar formation after wound healing is a major medical problem. A better understanding of the dynamic nuclear architecture of the genome during wound healing could provide insights into the underlying pathophysiology and enable novel therapeutic strategies. Here, we demonstrate that TGF-ß-induced fibrotic stress increases formation of the dynamic secondary DNA structures called G-quadruplexes in skin fibroblasts, which is coincident with increased expression of collagen 1. This G-quadruplex formation is attenuated by a small molecule inhibitor of intracellular Ca2+ influx and an anti-fibrotic compound. In addition, we identify G-quadruplex-forming sequences in the promoter region of COL1A1, which encodes collagen 1, and confirm their ability to form G-quadruplex structures under physiologically relevant conditions. Our findings reveal a link between G-quadruplexes and scar formation that may lead to novel therapeutic interventions.


Subject(s)
Collagen Type I/chemistry , Collagen Type I/genetics , Fibroblasts/drug effects , Transforming Growth Factor beta/pharmacology , Calcium/metabolism , Cells, Cultured , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis , G-Quadruplexes , Humans , Promoter Regions, Genetic , Transcription Initiation Site , Up-Regulation , Wound Healing
12.
Arch Biochem Biophys ; 665: 166-174, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30885674

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is a primary myocardial disorder, characterised by myocyte remodeling, disorganisation of sarcomeric proteins, impaired energy metabolism and altered cardiac contractility. Gene mutations encoding cardiac contractile proteins account for 60% of HCM aetiology. Current drug therapy including L-type calcium channel antagonists, are used to manage symptoms in patients with overt HCM, but no treatment exists that can reverse or prevent the cardiomyopathy. Design of effective drug therapy will require a clear understanding of the early pathophysiological mechanisms of the disease. Numerous studies have investigated specific aspects of HCM pathophysiology. This review brings these findings together, in order to develop a holistic understanding of the early pathophysiological mechanisms of the disease. We focus on gene mutations in cardiac myosin binding protein-C, ß-cardiac myosin heavy chain, cardiac troponin I, and cardiac troponin T, that comprise the majority of all HCM sarcomeric gene mutations. We find that although some similarities exist, each mutation leads to mutation-specific alterations in calcium handling, myofilament calcium sensitivity and mitochondrial metabolic function. This may contribute to the observed clinical phenotypic variability in sarcomeric-related HCM. An understanding of early mutation-specific mechanisms of the disease may provide useful markers of disease progression, and inform therapeutic design.


Subject(s)
Calcium/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Mitochondria, Heart/metabolism , Animals , Biomarkers/metabolism , Humans , Troponin T/metabolism
13.
Nat Commun ; 10(1): 1396, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30918256

ABSTRACT

Myoglobin, encoded by MB, is a small cytoplasmic globular hemoprotein highly expressed in cardiac myocytes and oxidative skeletal myofibers. Myoglobin binds O2, facilitates its intracellular transport and serves as a controller of nitric oxide and reactive oxygen species. Here, we identify a recurrent c.292C>T (p.His98Tyr) substitution in MB in fourteen members of six European families suffering from an autosomal dominant progressive myopathy with highly characteristic sarcoplasmic inclusions in skeletal and cardiac muscle. Myoglobinopathy manifests in adulthood with proximal and axial weakness that progresses to involve distal muscles and causes respiratory and cardiac failure. Biochemical characterization reveals that the mutant myoglobin has altered O2 binding, exhibits a faster heme dissociation rate and has a lower reduction potential compared to wild-type myoglobin. Preliminary studies show that mutant myoglobin may result in elevated superoxide levels at the cellular level. These data define a recognizable muscle disease associated with MB mutation.


Subject(s)
Inclusion Bodies/pathology , Muscle Fibers, Skeletal/pathology , Muscle Weakness/genetics , Muscular Diseases/genetics , Myocytes, Cardiac/pathology , Myoglobin/genetics , Adult , Female , Heart Failure/etiology , Heme/metabolism , Humans , Male , Middle Aged , Muscle Weakness/physiopathology , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/physiopathology , Muscular Diseases/diagnostic imaging , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Mutation , Oxygen/metabolism , Pedigree , Respiratory Insufficiency/etiology , Superoxides/metabolism , Tomography, X-Ray Computed , White People/genetics
14.
Sci Adv ; 5(12): eaay2118, 2019 12.
Article in English | MEDLINE | ID: mdl-31903419

ABSTRACT

Mammalian mitochondrial ribosomes are unique molecular machines that translate 11 leaderless mRNAs; however, it is not clear how mitoribosomes initiate translation, since mitochondrial mRNAs lack untranslated regions. Mitochondrial translation initiation shares similarities with prokaryotes, such as the formation of a ternary complex of fMet-tRNAMet, mRNA and the 28S subunit, but differs in the requirements for initiation factors. Mitochondria have two initiation factors: MTIF2, which closes the decoding center and stabilizes the binding of the fMet-tRNAMet to the leaderless mRNAs, and MTIF3, whose role is not clear. We show that MTIF3 is essential for survival and that heart- and skeletal muscle-specific loss of MTIF3 causes cardiomyopathy. We identify increased but uncoordinated mitochondrial protein synthesis in mice lacking MTIF3, resulting in loss of specific respiratory complexes. Ribosome profiling shows that MTIF3 is required for recognition and regulation of translation initiation of mitochondrial mRNAs and for coordinated assembly of OXPHOS complexes in vivo.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , Protein Biosynthesis/physiology , Animals , Cardiomyopathy, Dilated/genetics , Eukaryotic Initiation Factor-3/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism , RNA, Transfer, Met/metabolism , Ribosomes/metabolism
15.
JACC Basic Transl Sci ; 3(3): 391-402, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30062225

ABSTRACT

Current clinical trials demonstrate Duchenne muscular dystrophy (DMD) patients receiving phosphorodiamidate morpholino oligomer (PMO) therapy exhibit improved ambulation and stable pulmonary function; however, cardiac abnormalities remain. Utilizing the same PMO chemistry as current clinical trials, we have identified a non-toxic PMO treatment regimen that restores metabolic activity and prevents DMD cardiomyopathy. We propose that a treatment regimen of this nature may have the potential to significantly improve morbidity and mortality from DMD by improving ambulation, stabilizing pulmonary function, and preventing the development of cardiomyopathy.

16.
Sci Rep ; 8(1): 12538, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135446

ABSTRACT

Cell penetrating peptides (CPPs) offer great potential to deliver therapeutic molecules to previously inaccessible intracellular targets. However, many CPPs are inefficient and often leave their attached cargo stranded in the cell's endosome. We report a versatile platform for the isolation of peptides delivering a wide range of cargos into the cytoplasm of cells. We used this screening platform to identify multiple "Phylomer" CPPs, derived from bacterial and viral genomes. These peptides are amenable to conventional sequence optimization and engineering approaches for cell targeting and half-life extension. We demonstrate potent, functional delivery of protein, peptide, and nucleic acid analog cargos into cells using Phylomer CPPs. We validate in vivo activity in the cytoplasm, through successful transport of an oligonucleotide therapeutic fused to a Phylomer CPP in a disease model for Duchenne's muscular dystrophy. This report thus establishes a discovery platform for identifying novel, functional CPPs to expand the delivery landscape of druggable intracellular targets for biological therapeutics.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Drug Delivery Systems/methods , Drug Evaluation, Preclinical/methods , Animals , Bacteriophage T7 , Biotinylation , CHO Cells , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/toxicity , Circular Dichroism , Cricetulus , Disease Models, Animal , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Microscopy, Fluorescence , Muscular Dystrophy, Duchenne/drug therapy , Peptide Library , Repressor Proteins/genetics , Repressor Proteins/metabolism
17.
EMBO Rep ; 19(10)2018 10.
Article in English | MEDLINE | ID: mdl-30126926

ABSTRACT

The molecular roles of the dually targeted ElaC domain protein 2 (ELAC2) during nuclear and mitochondrial RNA processing in vivo have not been distinguished. We generated conditional knockout mice of ELAC2 to identify that it is essential for life and its activity is non-redundant. Heart and skeletal muscle-specific loss of ELAC2 causes dilated cardiomyopathy and premature death at 4 weeks. Transcriptome-wide analyses of total RNAs, small RNAs, mitochondrial RNAs, and miRNAs identified the molecular targets of ELAC2 in vivo We show that ELAC2 is required for processing of tRNAs and for the balanced maintenance of C/D box snoRNAs, miRNAs, and a new class of tRNA fragments. We identify that correct biogenesis of regulatory non-coding RNAs is essential for both cytoplasmic and mitochondrial protein synthesis and the assembly of mitochondrial ribosomes and cytoplasmic polysomes. We show that nuclear tRNA processing is required for the balanced production of snoRNAs and miRNAs for gene expression and that 3' tRNA processing is an essential step in the production of all mature mitochondrial RNAs and the majority of nuclear tRNAs.


Subject(s)
Endoribonucleases/genetics , Neoplasm Proteins/genetics , RNA, Mitochondrial/genetics , RNA, Untranslated/genetics , Animals , Cell Nucleus/genetics , Gene Expression Profiling , Mice , MicroRNAs/genetics , RNA, Small Nucleolar/genetics , RNA, Transfer/genetics , RNA, Untranslated/classification , RNA, Untranslated/isolation & purification
18.
Cell Rep ; 23(1): 127-142, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617655

ABSTRACT

The regulation of mitochondrial RNA life cycles and their roles in ribosome biogenesis and energy metabolism are not fully understood. We used CRISPR/Cas9 to generate heart- and skeletal-muscle-specific knockout mice of the pentatricopeptide repeat domain protein 1, PTCD1, and show that its loss leads to severe cardiomyopathy and premature death. Our detailed transcriptome-wide and functional analyses of these mice enabled us to identify the molecular role of PTCD1 as a 16S rRNA-binding protein essential for its stability, pseudouridylation, and correct biogenesis of the mitochondrial large ribosomal subunit. We show that impaired mitoribosome biogenesis can have retrograde signaling effects on nuclear gene expression through the transcriptional activation of the mTOR pathway and upregulation of cytoplasmic protein synthesis and pro-survival factors in the absence of mitochondrial translation. Taken together, our data show that impaired assembly of the mitoribosome exerts its consequences via differential regulation of mitochondrial and cytoplasmic protein synthesis.


Subject(s)
Mitochondrial Proteins/physiology , Mitochondrial Ribosomes/metabolism , Organelle Biogenesis , RNA, Ribosomal, 16S/metabolism , RNA-Binding Proteins/physiology , Animals , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Pseudouridine/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , TOR Serine-Threonine Kinases/metabolism
19.
ACS Sens ; 2(10): 1482-1490, 2017 10 27.
Article in English | MEDLINE | ID: mdl-28871791

ABSTRACT

Fibrotic diseases are among the most serious health issues with severe burdens due to their chronic nature and a large number of patients suffering from the debilitating effects and long-term sequelae. Collagenase treatment is a nonsurgical option but has limited results. To date, there is no potent noninvasive solution for fibrosis. Part of the reason for this is the lack of appropriate in vitro live cell screening tools to assess the efficacy of new therapeutical agents. Here, we demonstrate the utility of a cell-based electrochemical impedance biosensor platform to screen the efficacy of potential antifibrotic compounds. The platform employs a label-free and noninvasive strategy to detect the progression of fibrosis and the potency of the antifibrotic molecules in real-time. The fundamental principle that governs this novel system is that dynamic changes in cell shape and adhesion during fibrosis can be measured accurately by monitoring the changes in the impedance. This is achieved by growing the cells on a transparent interdigitated indium tin oxide (ITO) electrodes. It was demonstrated by monitoring the efficacy of a model antifibrotic compound, PXS64, on cells collected from patients with Dupuytren's contracture. We confirmed the validity of the developed biochemical impedance biosensor as an tool for in vitro screening of antifibrotic compounds and provided quantitative information on subcellular influences of the examined chemical molecules using correlative microscopy analyses that monitor the average cell area, cell morphology, and the amount and directionality of the deposited extracellular matrix protein collagen and measurement of cytosolic Ca2+ changes.


Subject(s)
Biosensing Techniques/methods , Dupuytren Contracture/drug therapy , Electric Impedance , Mannosides/pharmacology , Organophosphonates/pharmacology , Cells, Cultured , Dupuytren Contracture/metabolism , Electrodes , Humans , Tin Compounds/chemistry
20.
Sci Adv ; 3(8): e1700677, 2017 08.
Article in English | MEDLINE | ID: mdl-28835921

ABSTRACT

Mitochondrial gene expression is essential for energy production; however, an understanding of how it can influence physiology and metabolism is lacking. Several proteins from the pentatricopeptide repeat (PPR) family are essential for the regulation of mitochondrial gene expression, but the functions of the remaining members of this family are poorly understood. We created knockout mice to investigate the role of the PPR domain 1 (PTCD1) protein and show that loss of PTCD1 is embryonic lethal, whereas haploinsufficient, heterozygous mice develop age-induced obesity. The molecular defects and metabolic consequences of mitochondrial protein haploinsufficiency in vivo have not been investigated previously. We show that PTCD1 haploinsufficiency results in increased RNA metabolism, in response to decreased protein synthesis and impaired RNA processing that affect the biogenesis of the respiratory chain, causing mild uncoupling and changes in mitochondrial morphology. We demonstrate that with age, these effects lead to adult-onset obesity that results in liver steatosis and cardiac hypertrophy in response to tissue-specific differential regulation of the mammalian target of rapamycin pathways. Our findings indicate that changes in mitochondrial gene expression have long-term consequences on energy metabolism, providing evidence that haploinsufficiency of PTCD1 can be a major predisposing factor for the development of metabolic syndrome.


Subject(s)
Gene Expression Regulation , Genes, Mitochondrial , Genetic Association Studies , Genetic Predisposition to Disease , Obesity/genetics , Age of Onset , Animals , Disease Models, Animal , Energy Metabolism/genetics , Genotype , Glucose Intolerance , Hormones/metabolism , Insulin Resistance , Liver/metabolism , Liver/pathology , Liver/ultrastructure , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocardium/ultrastructure , Obesity/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL