Biochemistry and Pharmacology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 8 of 8
  • Item
    No Preview Available
    Integrated multi-omics for rapid rare disease diagnosis on a national scale
    Lunke, S ; Bouffler, SEE ; Patel, CVV ; Sandaradura, SAA ; Wilson, M ; Pinner, J ; Hunter, MFF ; Barnett, CPP ; Wallis, M ; Kamien, B ; Tan, TYY ; Freckmann, M-L ; Chong, B ; Phelan, D ; Francis, D ; Kassahn, KSS ; Ha, T ; Gao, S ; Arts, P ; Jackson, MRSR ; Scott, HSS ; Eggers, S ; Rowley, S ; Boggs, K ; Rakonjac, A ; Brett, GRR ; de Silva, MGG ; Springer, A ; Ward, M ; Stallard, K ; Simons, C ; Conway, T ; Halman, A ; Van Bergen, NJJ ; Sikora, T ; Semcesen, LNN ; Stroud, DAA ; Compton, AGG ; Thorburn, DRR ; Bell, KMM ; Sadedin, S ; North, KNN ; Christodoulou, J ; Stark, Z (NATURE PORTFOLIO, 2023-07)
    Critically ill infants and children with rare diseases need equitable access to rapid and accurate diagnosis to direct clinical management. Over 2 years, the Acute Care Genomics program provided whole-genome sequencing to 290 families whose critically ill infants and children were admitted to hospitals throughout Australia with suspected genetic conditions. The average time to result was 2.9 d and diagnostic yield was 47%. We performed additional bioinformatic analyses and transcriptome sequencing in all patients who remained undiagnosed. Long-read sequencing and functional assays, ranging from clinically accredited enzyme analysis to bespoke quantitative proteomics, were deployed in selected cases. This resulted in an additional 19 diagnoses and an overall diagnostic yield of 54%. Diagnostic variants ranged from structural chromosomal abnormalities through to an intronic retrotransposon, disrupting splicing. Critical care management changed in 120 diagnosed patients (77%). This included major impacts, such as informing precision treatments, surgical and transplant decisions and palliation, in 94 patients (60%). Our results provide preliminary evidence of the clinical utility of integrating multi-omic approaches into mainstream diagnostic practice to fully realize the potential of rare disease genomic testing in a timely manner.
  • Item
    Thumbnail Image
    Multi-omics identifies large mitoribosomal subunit instability caused by pathogenic MRPL39 variants as a cause of pediatric onset mitochondrial disease
    Amarasekera, SSC ; Hock, DH ; Lake, NJ ; Calvo, SE ; Gronborg, SW ; Krzesinski, E ; Amor, DJ ; Fahey, MC ; Simons, C ; Wibrand, F ; Mootha, VK ; Lek, M ; Lunke, S ; Stark, Z ; ostergaard, E ; Christodoulou, J ; Thorburn, DR ; Stroud, DA ; Compton, AG (OXFORD UNIV PRESS, 2023-07-20)
    MRPL39 encodes one of 52 proteins comprising the large subunit of the mitochondrial ribosome (mitoribosome). In conjunction with 30 proteins in the small subunit, the mitoribosome synthesizes the 13 subunits of the mitochondrial oxidative phosphorylation (OXPHOS) system encoded by mitochondrial Deoxyribonucleic acid (DNA). We used multi-omics and gene matching to identify three unrelated individuals with biallelic variants in MRPL39 presenting with multisystem diseases with severity ranging from lethal, infantile-onset (Leigh syndrome spectrum) to milder with survival into adulthood. Clinical exome sequencing of known disease genes failed to diagnose these patients; however quantitative proteomics identified a specific decrease in the abundance of large but not small mitoribosomal subunits in fibroblasts from the two patients with severe phenotype. Re-analysis of exome sequencing led to the identification of candidate single heterozygous variants in mitoribosomal genes MRPL39 (both patients) and MRPL15. Genome sequencing identified a shared deep intronic MRPL39 variant predicted to generate a cryptic exon, with transcriptomics and targeted studies providing further functional evidence for causation. The patient with the milder disease was homozygous for a missense variant identified through trio exome sequencing. Our study highlights the utility of quantitative proteomics in detecting protein signatures and in characterizing gene-disease associations in exome-unsolved patients. We describe Relative Complex Abundance analysis of proteomics data, a sensitive method that can identify defects in OXPHOS disorders to a similar or greater sensitivity to the traditional enzymology. Relative Complex Abundance has potential utility for functional validation or prioritization in many hundreds of inherited rare diseases where protein complex assembly is disrupted.
  • Item
    Thumbnail Image
    TEFM variants impair mitochondrial transcription causing childhood-onset neurological disease
    Van Haute, L ; O'Connor, E ; Diaz-Maldonado, H ; Munro, B ; Polavarapu, K ; Hock, DH ; Arunachal, G ; Athanasiou-Fragkouli, A ; Bardhan, M ; Barth, M ; Bonneau, D ; Brunetti-Pierri, N ; Cappuccio, G ; Caruana, NJ ; Dominik, N ; Goel, H ; Helman, G ; Houlden, H ; Lenaers, G ; Mention, K ; Murphy, D ; Nandeesh, B ; Olimpio, C ; Powell, CA ; Preethish-Kumar, V ; Procaccio, V ; Rius, R ; Rebelo-Guiomar, P ; Simons, C ; Vengalil, S ; Zaki, MS ; Ziegler, A ; Thorburn, DR ; Stroud, DA ; Maroofian, R ; Christodoulou, J ; Gustafsson, C ; Nalini, A ; Lochmueller, H ; Minczuk, M ; Horvath, R (NATURE PORTFOLIO, 2023-02-23)
    Mutations in the mitochondrial or nuclear genomes are associated with a diverse group of human disorders characterized by impaired mitochondrial respiration. Within this group, an increasing number of mutations have been identified in nuclear genes involved in mitochondrial RNA biology. The TEFM gene encodes the mitochondrial transcription elongation factor responsible for enhancing the processivity of mitochondrial RNA polymerase, POLRMT. We report for the first time that TEFM variants are associated with mitochondrial respiratory chain deficiency and a wide range of clinical presentations including mitochondrial myopathy with a treatable neuromuscular transmission defect. Mechanistically, we show muscle and primary fibroblasts from the affected individuals have reduced levels of promoter distal mitochondrial RNA transcripts. Finally, tefm knockdown in zebrafish embryos resulted in neuromuscular junction abnormalities and abnormal mitochondrial function, strengthening the genotype-phenotype correlation. Our study highlights that TEFM regulates mitochondrial transcription elongation and its defect results in variable, tissue-specific neurological and neuromuscular symptoms.
  • Item
    Thumbnail Image
    Biallelic Variants in PYROXD2 Cause a Severe Infantile Metabolic Disorder Affecting Mitochondrial Function
    Van Bergen, NJ ; Hock, DH ; Spencer, L ; Massey, S ; Stait, T ; Stark, Z ; Lunke, S ; Roesley, A ; Peters, H ; Lee, JY ; Le Fevre, A ; Heath, O ; Mignone, C ; Yang, JY-M ; Ryan, MM ; D'Arcy, C ; Nash, M ; Smith, S ; Caruana, NJ ; Thorburn, DR ; Stroud, DA ; White, SM ; Christodoulou, J ; Brown, NJ (MDPI, 2022-01)
    Pyridine Nucleotide-Disulfide Oxidoreductase Domain 2 (PYROXD2; previously called YueF) is a mitochondrial inner membrane/matrix-residing protein and is reported to regulate mitochondrial function. The clinical importance of PYROXD2 has been unclear, and little is known of the protein's precise biological function. In the present paper, we report biallelic variants in PYROXD2 identified by genome sequencing in a patient with suspected mitochondrial disease. The child presented with acute neurological deterioration, unresponsive episodes, and extreme metabolic acidosis, and received rapid genomic testing. He died shortly after. Magnetic resonance imaging (MRI) brain imaging showed changes resembling Leigh syndrome, one of the more common childhood mitochondrial neurological diseases. Functional studies in patient fibroblasts showed a heightened sensitivity to mitochondrial metabolic stress and increased mitochondrial superoxide levels. Quantitative proteomic analysis demonstrated decreased levels of subunits of the mitochondrial respiratory chain complex I, and both the small and large subunits of the mitochondrial ribosome, suggesting a mitoribosomal defect. Our findings support the critical role of PYROXD2 in human cells, and suggest that the biallelic PYROXD2 variants are associated with mitochondrial dysfunction, and can plausibly explain the child's clinical presentation.
  • Item
    No Preview Available
    Fatal Perinatal Mitochondrial Cardiac Failure Caused by Recurrent De Novo Duplications in the ATAD3 Locus
    Frazier, AE ; Compton, AG ; Kishita, Y ; Hock, DH ; Welch, AE ; Amarasekera, SSC ; Rius, R ; Formosa, LE ; Imai-Okazaki, A ; Francis, D ; Wang, M ; Lake, NJ ; Tregoning, S ; Jabbari, JS ; Lucattini, A ; Nitta, KR ; Ohtake, A ; Murayama, K ; Amor, DJ ; McGillivray, G ; Wong, FY ; van der Knaap, MS ; Vermeulen, RJ ; Wiltshire, EJ ; Fletcher, JM ; Lewis, B ; Baynam, G ; Ellaway, C ; Balasubramaniam, S ; Bhattacharya, K ; Freckmann, M-L ; Arbuckle, S ; Rodriguez, M ; Taft, RJ ; Sadedin, S ; Cowley, MJ ; Minoche, AE ; Calvo, SE ; Mootha, VK ; Ryan, MT ; Okazaki, Y ; Stroud, DA ; Simons, C ; Christodoulou, J ; Thorburn, DR (CELL PRESS, 2021-01-15)
    BACKGROUND: In about half of all patients with a suspected monogenic disease, genomic investigations fail to identify the diagnosis. A contributing factor is the difficulty with repetitive regions of the genome, such as those generated by segmental duplications. The ATAD3 locus is one such region, in which recessive deletions and dominant duplications have recently been reported to cause lethal perinatal mitochondrial diseases characterized by pontocerebellar hypoplasia or cardiomyopathy, respectively. METHODS: Whole exome, whole genome and long-read DNA sequencing techniques combined with studies of RNA and quantitative proteomics were used to investigate 17 subjects from 16 unrelated families with suspected mitochondrial disease. FINDINGS: We report six different de novo duplications in the ATAD3 gene locus causing a distinctive presentation including lethal perinatal cardiomyopathy, persistent hyperlactacidemia, and frequently corneal clouding or cataracts and encephalopathy. The recurrent 68 Kb ATAD3 duplications are identifiable from genome and exome sequencing but usually missed by microarrays. The ATAD3 duplications result in the formation of identical chimeric ATAD3A/ATAD3C proteins, altered ATAD3 complexes and a striking reduction in mitochondrial oxidative phosphorylation complex I and its activity in heart tissue. CONCLUSIONS: ATAD3 duplications appear to act in a dominant-negative manner and the de novo inheritance infers a low recurrence risk for families, unlike most pediatric mitochondrial diseases. More than 350 genes underlie mitochondrial diseases. In our experience the ATAD3 locus is now one of the five most common causes of nuclear-encoded pediatric mitochondrial disease but the repetitive nature of the locus means ATAD3 diagnoses may be frequently missed by current genomic strategies. FUNDING: Australian NHMRC, US Department of Defense, Japanese AMED and JSPS agencies, Australian Genomics Health Alliance and Australian Mito Foundation.
  • Item
    No Preview Available
    Abnormalities of mitochondrial dynamics and bioenergetics in neuronal cells from CDKL5 deficiency disorder
    Van Bergen, NJ ; Massey, S ; Stait, T ; Ellery, M ; Reljic, B ; Formosa, LE ; Quigley, A ; Dottori, M ; Thorburn, D ; Stroud, DA ; Christodoulou, J (ACADEMIC PRESS INC ELSEVIER SCIENCE, 2021-07)
    CDKL5 deficiency disorder (CDD) is a rare neurodevelopmental disorder caused by pathogenic variants in the Cyclin-dependent kinase-like 5 (CDKL5) gene, resulting in dysfunctional CDKL5 protein. It predominantly affects females and causes seizures in the first few months of life, ultimately resulting in severe intellectual disability. In the absence of targeted therapies, treatment is currently only symptomatic. CDKL5 is a serine/threonine kinase that is highly expressed in the brain, with a critical role in neuronal development. Evidence of mitochondrial dysfunction in CDD is gathering, but has not been studied extensively. We used human patient-derived induced pluripotent stem cells with a pathogenic truncating mutation (p.Arg59*) and CRISPR/Cas9 gene-corrected isogenic controls, differentiated into neurons, to investigate the impact of CDKL5 mutation on cellular function. Quantitative proteomics indicated mitochondrial defects in CDKL5 p.Arg59* neurons, and mitochondrial bioenergetics analysis confirmed decreased activity of mitochondrial respiratory chain complexes. Additionally, mitochondrial trafficking velocity was significantly impaired, and there was a higher percentage of stationary mitochondria. We propose mitochondrial dysfunction is contributing to CDD pathology, and should be a focus for development of targeted treatments for CDD.
  • Item
    Thumbnail Image
    Mutations in the exocyst component EXOC2 cause severe defects in human brain development
    Van Bergen, NJ ; Ahmed, SM ; Collins, F ; Cowley, M ; Vetro, A ; Dale, RC ; Hock, DH ; de Caestecker, C ; Menezes, M ; Massey, S ; Ho, G ; Pisano, T ; Glover, S ; Gusman, J ; Stroud, DA ; Dinger, M ; Guerrini, R ; Macara, IG ; Christodoulou, J (ROCKEFELLER UNIV PRESS, 2020-10)
    The exocyst, an octameric protein complex, is an essential component of the membrane transport machinery required for tethering and fusion of vesicles at the plasma membrane. We report pathogenic variants in an exocyst subunit, EXOC2 (Sec5). Affected individuals have severe developmental delay, dysmorphism, and brain abnormalities; variability associated with epilepsy; and poor motor skills. Family 1 had two offspring with a homozygous truncating variant in EXOC2 that leads to nonsense-mediated decay of EXOC2 transcript, a severe reduction in exocytosis and vesicle fusion, and undetectable levels of EXOC2 protein. The patient from Family 2 had a milder clinical phenotype and reduced exocytosis. Cells from both patients showed defective Arl13b localization to the primary cilium. The discovery of mutations that partially disable exocyst function provides valuable insight into this essential protein complex in neural development. Since EXOC2 and other exocyst complex subunits are critical to neuronal function, our findings suggest that EXOC2 variants are the cause of the patients' neurological disorders.
  • Item
    Thumbnail Image
    Multiomic analysis elucidates Complex I deficiency caused by a deep intronic variant in NDUFB10
    Helman, G ; Compton, AG ; Hock, DH ; Walkiewicz, M ; Brett, GR ; Pais, L ; Tan, TY ; De Paoli-Iseppi, R ; Clark, MB ; Christodoulou, J ; White, SM ; Thorburn, DR ; Stroud, DA ; Stark, Z ; Simons, C (WILEY-HINDAWI, 2021-01)
    The diagnosis of Mendelian disorders following uninformative exome and genome sequencing remains a challenging and often unmet need. Following uninformative exome and genome sequencing of a family quartet including two siblings with suspected mitochondrial disorder, RNA sequencing (RNAseq) was pursued in one sibling. Long-read amplicon sequencing was used to determine and quantify transcript structure. Immunoblotting studies and quantitative proteomics were performed to demonstrate functional impact. Differential expression analysis of RNAseq data identified significantly decreased expression of the mitochondrial OXPHOS Complex I subunit NDUFB10 associated with a cryptic exon in intron 1 of NDUFB10, that included an in-frame stop codon. The cryptic exon contained a rare intronic variant that was homozygous in both affected siblings. Immunoblot and quantitative proteomic analysis of fibroblasts revealed decreased abundance of Complex I subunits, providing evidence of isolated Complex I deficiency. Through multiomic analysis we present data implicating a deep intronic variant in NDUFB10 as the cause of mitochondrial disease in two individuals, providing further support of the gene-disease association. This study highlights the importance of transcriptomic and proteomic analyses as complementary diagnostic tools in patients undergoing genome-wide diagnostic evaluation.