72 results on '"Sarah E. Calvo"'
Search Results
2. Multi-omics identifies large mitoribosomal subunit instability caused by pathogenic MRPL39 variants as a cause of pediatric onset mitochondrial disease
- Author
-
Sumudu S C Amarasekera, Daniella H Hock, Nicole J Lake, Sarah E Calvo, Sabine W Grønborg, Emma I Krzesinski, David J Amor, Michael C Fahey, Cas Simons, Flemming Wibrand, Vamsi K Mootha, Monkol Lek, Sebastian Lunke, Zornitza Stark, Elsebet Østergaard, John Christodoulou, David R Thorburn, David A Stroud, and Alison G Compton
- Subjects
Genetics ,General Medicine ,Molecular Biology ,Genetics (clinical) - Abstract
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 or OXPHOS system encoded by mitochondrial 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 milder disease was homozygous for a missense variant identified through trio exome sequencing. Our study highlights the utility of quantitative proteomics in detection of protein signatures and in characterization of 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.
- Published
- 2023
3. Mitochondrial DNA variation across 56,434 individuals in gnomAD
- Author
-
Kristen M, Laricchia, Nicole J, Lake, Nicholas A, Watts, Megan, Shand, Andrea, Haessly, Laura, Gauthier, David, Benjamin, Eric, Banks, Jose, Soto, Kiran, Garimella, James, Emery, Heidi L, Rehm, Daniel G, MacArthur, Grace, Tiao, Monkol, Lek, Vamsi K, Mootha, and Sarah E, Calvo
- Subjects
Cell Nucleus ,Genome ,Gene Frequency ,Genome, Mitochondrial ,Genetics ,Humans ,Sequence Analysis, DNA ,DNA, Mitochondrial ,Genetics (clinical) ,Mitochondria - Abstract
Genomic databases of allele frequency are extremely helpful for evaluating clinical variants of unknown significance; however, until now, databases such as the Genome Aggregation Database (gnomAD) have focused on nuclear DNA and have ignored the mitochondrial genome (mtDNA). Here, we present a pipeline to call mtDNA variants that addresses three technical challenges: (1) detecting homoplasmic and heteroplasmic variants, present, respectively, in all or a fraction of mtDNA molecules; (2) circular mtDNA genome; and (3) misalignment of nuclear sequences of mitochondrial origin (NUMTs). We observed that mtDNA copy number per cell varied across gnomAD cohorts and influenced the fraction of NUMT-derived false-positive variant calls, which can account for the majority of putative heteroplasmies. To avoid false positives, we excluded contaminated samples, cell lines, and samples prone to NUMT misalignment due to few mtDNA copies. Furthermore, we report variants with heteroplasmy ≥10%. We applied this pipeline to 56,434 whole-genome sequences in the gnomAD v3.1 database that includes individuals of European (58%), African (25%), Latino (10%), and Asian (5%) ancestry. Our gnomAD v3.1 release contains population frequencies for 10,850 unique mtDNA variants at more than half of all mtDNA bases. Importantly, we report frequencies within each nuclear ancestral population and mitochondrial haplogroup. Homoplasmic variants account for most variant calls (98%) and unique variants (85%). We observed that 1/250 individuals carry a pathogenic mtDNA variant with heteroplasmy above 10%. These mtDNA population allele frequencies are freely accessible and will aid in diagnostic interpretation and research studies.
- Published
- 2022
4. Nuclear genetic control of mtDNA copy number and heteroplasmy in humans
- Author
-
Rahul Gupta, Masahiro Kanai, Timothy J. Durham, Kristin Tsuo, Jason G. McCoy, Patrick F. Chinnery, Konrad J. Karczewski, Sarah E. Calvo, Benjamin M. Neale, and Vamsi K. Mootha
- Subjects
Article - Abstract
Human mitochondria contain a high copy number, maternally transmitted genome (mtDNA) that encodes 13 proteins required for oxidative phosphorylation. Heteroplasmy arises when multiple mtDNA variants co-exist in an individual and can exhibit complex dynamics in disease and in aging. As all proteins involved in mtDNA replication and maintenance are nuclear-encoded, heteroplasmy levels can, in principle, be under nuclear genetic control, however this has never been shown in humans. Here, we develop algorithms to quantify mtDNA copy number (mtCN) and heteroplasmy levels using blood-derived whole genome sequences from 274,832 individuals of diverse ancestry and perform GWAS to identify nuclear loci controlling these traits. After careful correction for blood cell composition, we observe that mtCN declines linearly with age and is associated with 92 independent nuclear genetic loci. We find that nearly every individual carries heteroplasmic variants that obey two key patterns: (1) heteroplasmic single nucleotide variants are somatic mutations that accumulate sharply after age 70, while (2) heteroplasmic indels are maternally transmitted as mtDNA mixtures with resulting levels influenced by 42 independent nuclear loci involved in mtDNA replication, maintenance, and novel pathways. These nuclear loci do not appear to act by mtDNA mutagenesis, but rather, likely act by conferring a replicative advantage to specific mtDNA molecules. As an illustrative example, the most common heteroplasmy we identify is a length variant carried by >50% of humans at position m.302 within a G-quadruplex known to serve as a replication switch. We find that this heteroplasmic variant exertscis-acting genetic control over mtDNA abundance and is itself undertrans-acting genetic control of nuclear loci encoding protein components of this regulatory switch. Our study showcases how nuclear haplotype can privilege the replication of specific mtDNA molecules to shape mtCN and heteroplasmy dynamics in the human population.
- Published
- 2023
5. CLIC, a tool for expanding biological pathways based on co-expression across thousands of datasets.
- Author
-
Yang Li, Alexis A Jourdain, Sarah E Calvo, Jun S Liu, and Vamsi K Mootha
- Subjects
Biology (General) ,QH301-705.5 - Abstract
In recent years, there has been a huge rise in the number of publicly available transcriptional profiling datasets. These massive compendia comprise billions of measurements and provide a special opportunity to predict the function of unstudied genes based on co-expression to well-studied pathways. Such analyses can be very challenging, however, since biological pathways are modular and may exhibit co-expression only in specific contexts. To overcome these challenges we introduce CLIC, CLustering by Inferred Co-expression. CLIC accepts as input a pathway consisting of two or more genes. It then uses a Bayesian partition model to simultaneously partition the input gene set into coherent co-expressed modules (CEMs), while assigning the posterior probability for each dataset in support of each CEM. CLIC then expands each CEM by scanning the transcriptome for additional co-expressed genes, quantified by an integrated log-likelihood ratio (LLR) score weighted for each dataset. As a byproduct, CLIC automatically learns the conditions (datasets) within which a CEM is operative. We implemented CLIC using a compendium of 1774 mouse microarray datasets (28628 microarrays) or 1887 human microarray datasets (45158 microarrays). CLIC analysis reveals that of 910 canonical biological pathways, 30% consist of strongly co-expressed gene modules for which new members are predicted. For example, CLIC predicts a functional connection between protein C7orf55 (FMC1) and the mitochondrial ATP synthase complex that we have experimentally validated. CLIC is freely available at www.gene-clic.org. We anticipate that CLIC will be valuable both for revealing new components of biological pathways as well as the conditions in which they are active.
- Published
- 2017
- Full Text
- View/download PDF
6. Effectors enabling adaptation to mitochondrial complex I loss in Hürthle cell carcinoma
- Author
-
Raj K. Gopal, Venkata R. Vantaku, Apekshya Panda, Bryn Reimer, Sneha Rath, Tsz-Leung To, Adam S. Fisch, Murat Cetinbas, Maia Livneh, Michael J. Calcaterra, Benjamin J. Gigliotti, Kerry Pierce, Clary B. Clish, Dora Dias-Santagata, Peter M. Sadow, Lori J. Wirth, Gilbert H. Daniels, Ruslan I. Sadreyev, Sarah E. Calvo, Sareh Parangi, and Vamsi K. Mootha
- Abstract
SummaryOncocytic (Hürthle cell) carcinoma of the thyroid (HCC) is genetically characterized by complex I mitochondrial DNA mutations and widespread chromosomal losses. Here, we utilize RNA-seq and metabolomics to identify candidate molecular effectors activated by these genetic drivers. We find glutathione biosynthesis, amino acid metabolism, mitochondrial unfolded protein response, and lipid peroxide scavenging, a safeguard against ferroptosis, to be increased in HCC. A CRISPR-Cas9 knockout screen in a new HCC model reveals which pathways are key for fitness, and highlights loss of GPX4, a defense against ferroptosis, as a strong liability. Rescuing complex I redox activity with the yeast NADH dehydrogenase (NDI1) in HCC cells diminishes ferroptosis sensitivity, while inhibiting complex I in normal thyroid cells augments ferroptosis induction. Our work demonstrates unmitigated lipid peroxide stress to be an HCC vulnerability that is mechanistically coupled to the genetic loss of mitochondrial complex I activity.SignificanceOncocytic (Hürthle cell) carcinoma of the thyroid (HCC) is a unique tumor with a remarkable accumulation of mitochondria. HCC harbors unique genetic alterations, including mitochondrial DNA (mtDNA) mutations in complex I genes and widespread loss-of-heterozygosity in the nuclear DNA. With less favorable clinical outcomes, new therapies for HCC are needed, especially since these tumors show intrinsic resistance to radioactive iodine, which is one of the main treatments for metastatic well-differentiated thyroid cancer. An absence of authentic HCC cell lines and animal models has hindered the mechanistic understanding of this disease and slowed therapeutic progress. In this study, we describe the transcriptomic and metabolomic landscape of HCC and present new HCC models that recapitulate key mtDNA and nuclear DNA alterations. A targeted CRISPR-Cas9 knockout screen in an HCC cell line highlights the molecular programs nominated by our -omics profiling that are required for cell fitness. This screen suggests that lipid peroxide scavenging, a defense system against an iron-dependent form of cell death known as ferroptosis, is a vulnerability in HCC that is coupled to complex I loss, and that targeting this pathway may help patients with HCC.
- Published
- 2022
7. MitoCarta3.0: an updated mitochondrial proteome now with sub-organelle localization and pathway annotations
- Author
-
Tslil Ast, Shayan Sadre, Pallavi R. Joshi, Owen S. Skinner, Anna V. Kotrys, Melissa A. Walker, Tsz-Leung To, Zenon Grabarek, Hong Wang, Maria Miranda, Wendy H. W. Hung, Joshua D. Meisel, Mary E. Haas, Alexis A. Jourdain, Sneha Rath, Connie Chan, Sharon H. Kim, Chen-Ching Yuan, Hardik Shah, Patrick S. Ward, Timothy J. Durham, Robert S. Rogers, Anupam Patgiri, Rohit Sharma, Russell P. Goodman, Stephanie S Lam, Rahul Gupta, Jason G. McCoy, Sarah E. Calvo, Apekshya Panda, Vamsi K. Mootha, and Jordan Wengrod
- Subjects
Mitochondrial DNA ,Nuclear gene ,Proteome ,AcademicSubjects/SCI00010 ,Datasets as Topic ,Computational biology ,Mitochondrion ,Biology ,DNA, Mitochondrial ,Mass Spectrometry ,Machine Learning ,Mitochondrial Proteins ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,Database Issue ,Animals ,Humans ,Databases, Protein ,Gene ,030304 developmental biology ,Internet ,0303 health sciences ,Bayes Theorem ,Molecular Sequence Annotation ,Mitochondria ,Mitochondrial Membranes ,Human genome ,Intermembrane space ,Software ,030217 neurology & neurosurgery ,Organelle localization - Abstract
The mammalian mitochondrial proteome is under dual genomic control, with 99% of proteins encoded by the nuclear genome and 13 originating from the mitochondrial DNA (mtDNA). We previously developed MitoCarta, a catalogue of over 1000 genes encoding the mammalian mitochondrial proteome. This catalogue was compiled using a Bayesian integration of multiple sequence features and experimental datasets, notably protein mass spectrometry of mitochondria isolated from fourteen murine tissues. Here, we introduce MitoCarta3.0. Beginning with the MitoCarta2.0 inventory, we performed manual review to remove 100 genes and introduce 78 additional genes, arriving at an updated inventory of 1136 human genes. We now include manually curated annotations of sub-mitochondrial localization (matrix, inner membrane, intermembrane space, outer membrane) as well as assignment to 149 hierarchical ‘MitoPathways’ spanning seven broad functional categories relevant to mitochondria. MitoCarta3.0, including sub-mitochondrial localization and MitoPathway annotations, is freely available at http://www.broadinstitute.org/mitocarta and should serve as a continued community resource for mitochondrial biology and medicine.
- Published
- 2020
8. Combinatorial G x G x E CRISPR screening and functional analysis highlights SLC25A39 in mitochondrial GSH transport
- Author
-
Bryn Reinstadler, Vamsi K. Mootha, Hardik Shah, Katie Byrne, Hongying Shen, Xiaojian Shi, Olga Goldberger, Tsz-Leung To, John G. Doench, Luanna Summer, and Sarah E. Calvo
- Subjects
chemistry.chemical_compound ,chemistry ,Biochemistry ,Galactose ,Organelle ,Mutagenesis ,CRISPR ,Transporter ,Glutathione ,Oxidative phosphorylation ,Gene - Abstract
The SLC25 carrier family consists of 53 transporters that shuttle nutrients and co-factors across mitochondrial membranes1-3. The family is highly redundant and their transport activities are coupled to metabolic state. Here, we introduce a pooled, dual CRISPR screening strategy that knocks out pairs of transporters in four metabolic states — glucose, galactose, OXPHOS inhibition, and absence of pyruvate — designed to unmask the inter-dependence of these genes. In total, we screened 63 genes in four metabolic states, corresponding to 2016 single and pair-wise genetic perturbations. We recovered 19 gene-by-environment (GxE) interactions and 9 gene-by-gene (GxG) interactions. One GxE interaction hit illustrated that the fitness defect in the mitochondrial folate carrier (SLC25A32) KO cells were genetically buffered in galactose due to a lack of substrate in de novo purine biosynthesis. Another GxE interaction hit revealed non-equivalence of the paralogous ATP/ADP exchangers (ANTs) with ANT2 specifically required during OXPHOS inhibition. GxG analysis highlighted a buffering interaction between the iron transporter SLC25A37 and the poorly characterized SLC25A39. Mitochondrial metabolite profiling, organelle transport assays, and structure-guided mutagenesis suggest SLC25A39 is critical for mitochondrial glutathione (GSH) transport. Our work underscores the importance of systematically investigating family-wide genetic interactions between mitochondrial transporters across many metabolic environments.
- Published
- 2021
9. Combinatorial G x G x E CRISPR screening and functional analysis highlights SLC25A39 in mitochondrial GSH transport
- Author
-
Xiaojian Shi, Bryn Reinstadler, Hardik Shah, Tsz-Leung To, Katie Byrne, Luanna Summer, Sarah E. Calvo, Olga Goldberger, John G. Doench, Vamsi K. Mootha, and Hongying Shen
- Abstract
The SLC25 carrier family consists of 53 transporters that shuttle nutrients and co-factors across mitochondrial membranes1-3. The family is highly redundant and their transport activities coupled to metabolic state. Here, we introduce a pooled, dual CRISPR screening strategy that knocks out pairs of transporters in four metabolic states — glucose, galactose, OXPHOS inhibition, and absence of pyruvate — designed to unmask the inter-dependence of these genes. In total, we screened 63 genes in four metabolic states, corresponding to 2016 single and pair-wise genetic perturbations. We recovered 19 gene-by-environment (GxE) interactions and 9 gene-by-gene (GxG) interactions. One GxE interaction hit illustrated that the fitness defect in the mitochondrial folate carrier (SLC25A32) KO cells was genetically buffered in galactose due to a lack of substrate in de novo purine biosynthesis. Another GxE interaction hit revealed non-equivalence of the paralogous ATP/ADP exchangers (ANTs) with ANT2 specifically required during OXPHOS inhibition. GxG analysis highlighted a buffering interaction between the iron transporter SLC25A37 and the poorly characterized SLC25A39. Mitochondrial metabolite profiling, organelle transport assays, and structure-guided mutagenesis suggests SLC25A39 is critical for mitochondrial glutathione (GSH) transport. Our work underscores the importance of systemetically investigating family-wide genetic interactions between mitochondrial transporters across many metabolic environments.
- Published
- 2021
10. Mitochondrial DNA variation across 56,434 individuals in gnomAD
- Author
-
Nicholas A. Watts, Monkol Lek, James Emery, Sarah E. Calvo, Grace Tiao, Daniel G. MacArthur, Eric Banks, Laura D. Gauthier, Vamsi K. Mootha, Andrea Haessly, David Benjamin, Megan Shand, Jose Soto, Heidi L. Rehm, Nicole J. Lake, Kristen M. Laricchia, and Kiran V. Garimella
- Subjects
Genetics ,Mitochondrial DNA ,education.field_of_study ,Unknown Significance ,Population ,Numt ,Biology ,education ,Allele frequency ,Genome ,Heteroplasmy ,Human mitochondrial DNA haplogroup - Abstract
Databases of allele frequency are extremely helpful for evaluating clinical variants of unknown significance; however, until now, genetic databases such as the Genome Aggregation Database (gnomAD) have ignored the mitochondrial genome (mtDNA). Here we present a pipeline to call mtDNA variants that addresses three technical challenges: (i) detecting homoplasmic and heteroplasmic variants, present respectively in all or a fraction of mtDNA molecules, (ii) circular mtDNA genome, and (iii) misalignment of nuclear sequences of mitochondrial origin (NUMTs). We observed that mtDNA copy number per cell varied across gnomAD cohorts and influenced the fraction of NUMT-derived false-positive variant calls, which can account for the majority of putative heteroplasmies. To avoid false positives, we excluded samples prone to NUMT misalignment (few mtDNA copies per cell), cell line artifacts (many mtDNA copies per cell), or with contamination and we reported variants with heteroplasmy greater than 10%. We applied this pipeline to 56,434 whole genome sequences in the gnomAD v3.1 database that includes individuals of European (58%), African (25%), Latino (10%), and Asian (5%) ancestry. Our gnomAD v3.1 release contains population frequencies for 10,850 unique mtDNA variants at more than half of all mtDNA bases. Importantly, we report frequencies within each nuclear ancestral population and mitochondrial haplogroup. Homoplasmic variants account for most variant calls (98%) and unique variants (85%). We observed that 1/250 individuals carry a pathogenic mtDNA variant with heteroplasmy above 10%. These mitochondrial population allele frequencies are publicly available at gnomad.broadinstitute.org and will aid in diagnostic interpretation and research studies.
- Published
- 2021
11. Loss of LUC7L2 and U1 snRNP subunits shifts energy metabolism from glycolysis to OXPHOS
- Author
-
Bridget E. Begg, Eran Mick, Hardik Shah, Steven M. Blue, Alexis A. Jourdain, Owen S. Skinner, Rohit Sharma, Sarah E. Calvo, Gene W. Yeo, Vamsi K. Mootha, and Christopher B. Burge
- Subjects
0303 health sciences ,Spliceosome ,biology ,Cell Biology ,Oxidative phosphorylation ,SLC7A11 ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,PFKM ,7q ,LUC7 ,MDS ,Tarui disease ,cancer ,ferroptosis ,myelodysplastic syndrome ,phosphofructokinase ,spliceosome ,system X(c)(−) ,RNA splicing ,biology.protein ,snRNP ,Glycolysis ,Molecular Biology ,030217 neurology & neurosurgery ,030304 developmental biology ,Phosphofructokinase - Abstract
Oxidative phosphorylation (OXPHOS) and glycolysis are the two major pathways for ATP production. The reliance on each varies across tissues and cell states, and can influence susceptibility to disease. At present, the full set of molecular mechanisms governing the relative expression and balance of these two pathways is unknown. Here, we focus on genes whose loss leads to an increase in OXPHOS activity. Unexpectedly, this class of genes is enriched for components of the pre-mRNA splicing machinery, in particular for subunits of the U1 snRNP. Among them, we show that LUC7L2 represses OXPHOS and promotes glycolysis by multiple mechanisms, including (1) splicing of the glycolytic enzyme PFKM to suppress glycogen synthesis, (2) splicing of the cystine/glutamate antiporter SLC7A11 (xCT) to suppress glutamate oxidation, and (3) secondary repression of mitochondrial respiratory supercomplex formation. Our results connect LUC7L2 expression and, more generally, the U1 snRNP to cellular energy metabolism.
- Published
- 2021
12. Fatal perinatal mitochondrial cardiac failure caused by recurrent
- Author
-
Ann E, Frazier, Alison G, Compton, Yoshihito, Kishita, Daniella H, Hock, AnneMarie E, Welch, Sumudu S C, Amarasekera, Rocio, Rius, Luke E, Formosa, Atsuko, Imai-Okazaki, David, Francis, Min, Wang, Nicole J, Lake, Simone, Tregoning, Jafar S, Jabbari, Alexis, Lucattini, Kazuhiro R, Nitta, Akira, Ohtake, Kei, Murayama, David J, Amor, George, McGillivray, Flora Y, Wong, Marjo S, van der Knaap, R, Jeroen Vermeulen, Esko J, Wiltshire, Janice M, Fletcher, Barry, Lewis, Gareth, Baynam, Carolyn, Ellaway, Shanti, Balasubramaniam, Kaustuv, Bhattacharya, Mary-Louise, Freckmann, Susan, Arbuckle, Michael, Rodriguez, Ryan J, Taft, Simon, Sadedin, Mark J, Cowley, André E, Minoche, Sarah E, Calvo, Vamsi K, Mootha, Michael T, Ryan, Yasushi, Okazaki, David A, Stroud, Cas, Simons, John, Christodoulou, and David R, Thorburn
- Subjects
Heart Failure ,Mitochondrial Proteins ,Mitochondrial Diseases ,Australia ,ATPases Associated with Diverse Cellular Activities ,Humans ,Membrane Proteins ,Cardiomyopathies ,Child ,United States - Abstract
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. TheWhole 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.We report six differentAustralian NHMRC, US Department of Defense, Japanese AMED and JSPS agencies, Australian Genomics Health Alliance and Australian Mito Foundation.
- Published
- 2021
13. Fatal Perinatal Mitochondrial Cardiac Failure Caused by Recurrent De Novo Duplications in the ATAD3 Locus
- Author
-
Flora Y. Wong, Nicole J. Lake, Michael T. Ryan, Anne Marie E. Welch, Kazuhiro R. Nitta, Sarah E. Calvo, Yoshihito Kishita, Daniella H Hock, David A. Stroud, Simone Tregoning, Gareth Baynam, Michael Rodriguez, André E. Minoche, Susan Arbuckle, Kaustuv Bhattacharya, Yasushi Okazaki, David J. Amor, Akira Ohtake, George McGillivray, John Christodoulou, R. Jeroen Vermeulen, Mary Louise Freckmann, Atsuko Imai-Okazaki, Shanti Balasubramaniam, Carolyn Ellaway, Luke E. Formosa, David R. Thorburn, Marjo S van der Knaap, Alison G. Compton, Rocio Rius, Janice M. Fletcher, Mark J. Cowley, Cas Simons, Ann E. Frazier, Kei Murayama, Alexis Lucattini, Ryan J. Taft, Barry Lewis, David Francis, Simon Sadedin, Sumudu S.C. Amarasekera, Jafar S. Jabbari, Vamsi K. Mootha, Min Wang, Esko Wiltshire, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, Pediatric surgery, Functional Genomics, Klinische Neurowetenschappen, MUMC+: MA Med Staf Spec Neurologie (9), and RS: MHeNs - R1 - Cognitive Neuropsychiatry and Clinical Neuroscience
- Subjects
quantitative proteomics ,COMPUTATIONAL PLATFORM ,Translation to Patients ,Mitochondrial disease ,Pontocerebellar hypoplasia ,segmental duplication ,Locus (genetics) ,Biology ,HIGH-THROUGHPUT ,Genome ,GENOMIC DISORDERS ,SDG 3 - Good Health and Well-being ,medicine ,de novo duplication ,genomics ,Copy-number variation ,COPY-NUMBER VARIANTS ,ATAD3 ,Exome sequencing ,Segmental duplication ,Genetics ,ARCHITECTURE ,MEMBRANE-PROTEIN ,MUTATIONS ,CHOLESTEROL ,General Medicine ,DNA ,medicine.disease ,COMPLEX I DEFICIENCY ,mitochondrial disease ,perinatal death ,Human genome ,cardiomyopathy - Abstract
Summary 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 6 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, US National Institutes of Health, Japanese AMED and JSPS agencies, Australian Genomics Health Alliance, and Australian Mito Foundation.
- Published
- 2021
14. The Proteomic Signature of Septic Shock Differs from Cardiogenic Shock or Bacteremia Without Sepsis or Shock
- Author
-
Kathryn A. Hibbert, Sarah E. Calvo, Rohit Sharma, Robert S. Rogers, N.A. Pulido, Vamsi K. Mootha, Kelsey Brait, and B.T.T. Thompson
- Subjects
Sepsis ,medicine.medical_specialty ,Septic shock ,business.industry ,Internal medicine ,Shock (circulatory) ,Bacteremia ,Cardiogenic shock ,medicine ,Cardiology ,medicine.symptom ,medicine.disease ,business - Published
- 2020
15. Genetic screen for cell fitness in high or low oxygen highlights mitochondrial and lipid metabolism
- Author
-
Owen S. Skinner, Sarah E. Calvo, Tslil Ast, Vamsi K. Mootha, Tsz-Leung To, Andrew L. Markhard, and Isha H. Jain
- Subjects
chemistry.chemical_element ,Biology ,Oxygen ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Oxygen homeostasis ,Humans ,Genetic Testing ,Hypoxia ,Gene ,Gene knockout ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Reactive oxygen species ,Lipid metabolism ,Peroxisome ,Lipid Metabolism ,Lipids ,Cell Hypoxia ,Cell biology ,Mitochondria ,HEK293 Cells ,chemistry ,K562 Cells ,Reactive Oxygen Species ,Transcriptome ,030217 neurology & neurosurgery ,Genetic screen ,Genome-Wide Association Study ,Signal Transduction - Abstract
Summary Human cells are able to sense and adapt to variations in oxygen levels. Historically, much research in this field has focused on hypoxia-inducible factor (HIF) signaling and reactive oxygen species (ROS). Here, we perform genome-wide CRISPR growth screens at 21%, 5%, and 1% oxygen to systematically identify gene knockouts with relative fitness defects in high oxygen (213 genes) or low oxygen (109 genes), most without known connection to HIF or ROS. Knockouts of many mitochondrial pathways thought to be essential, including complex I and enzymes in Fe-S biosynthesis, grow relatively well at low oxygen and thus are buffered by hypoxia. In contrast, in certain cell types, knockout of lipid biosynthetic and peroxisomal genes causes fitness defects only in low oxygen. Our resource nominates genetic diseases whose severity may be modulated by oxygen and links hundreds of genes to oxygen homeostasis.
- Published
- 2020
16. Spatiotemporal compartmentalization of hepatic NADH and NADPH metabolism
- Author
-
Vamsi K. Mootha, Russell P. Goodman, and Sarah E. Calvo
- Subjects
0301 basic medicine ,Mitochondrion ,Biochemistry ,Redox ,03 medical and health sciences ,Cytosol ,Spatio-Temporal Analysis ,medicine ,Animals ,Homeostasis ,Humans ,Molecular Biology ,chemistry.chemical_classification ,Thematic Minireviews ,Fatty liver ,Cell Biology ,Metabolism ,Compartmentalization (fire protection) ,NAD ,medicine.disease ,Mitochondria ,Oxidative Stress ,030104 developmental biology ,Enzyme ,Liver ,chemistry ,NAD+ kinase ,Energy Metabolism ,Oxidation-Reduction ,Metabolic Networks and Pathways ,NADP - Abstract
Compartmentalization is a fundamental design principle of eukaryotic metabolism. Here, we review the compartmentalization of NAD(+)/NADH and NADP(+)/NADPH with a focus on the liver, an organ that experiences the extremes of biochemical physiology each day. Historical studies of the liver, using classical biochemical fractionation and measurements of redox-coupled metabolites, have given rise to the prevailing view that mitochondrial NAD(H) pools tend to be oxidized and important for energy homeostasis, whereas cytosolic NADP(H) pools tend to be highly reduced for reductive biosynthesis. Despite this textbook view, many questions still remain as to the relative size of these subcellular pools and their redox ratios in different physiological states, and to what extent such redox ratios are simply indicators versus drivers of metabolism. By performing a bioinformatic survey, we find that the liver expresses 352 known or predicted enzymes composing the hepatic NAD(P)ome, i.e. the union of all predicted enzymes producing or consuming NADP(H) or NAD(H) or using them as a redox co-factor. Notably, less than half are predicted to be localized within the cytosol or mitochondria, and a very large fraction of these genes exhibit gene expression patterns that vary during the time of day or in response to fasting or feeding. A future challenge lies in applying emerging new genetic tools to measure and manipulate in vivo hepatic NADP(H) and NAD(H) with subcellular and temporal resolution. Insights from such fundamental studies will be crucial in deciphering the pathogenesis of very common diseases known to involve alterations in hepatic NAD(P)H, such as diabetes and fatty liver disease.
- Published
- 2018
17. Biallelic Mutations in MRPS34 Lead to Instability of the Small Mitoribosomal Subunit and Leigh Syndrome
- Author
-
Agnès Rötig, Marlène Rio, Vamsi K. Mootha, Zhancheng Zhang, Nicole J. Lake, Benedetta Ruzzenente, David A. Stroud, Nathalie Bodaert, Elizabeth M. McCormick, Tara R. Richman, Zarazuela Zolkipli-Cunningham, Sander M. Houten, Marni J. Falk, Kyle Retterer, Alison G. Compton, Mingma D. Sherpa, Metodi D. Metodiev, James Byrnes, Katrina Haude, Zahra Assouline, Hayley S. Mountford, Juliette Pulman, Aleksandra Filipovska, John Christodoulou, Ingrid Cristian, Eric E. Schadt, Renkui Bai, Bryn D. Webb, Sarah E. Calvo, David R. Thorburn, Coralie Zangarelli, and Laboratory Genetic Metabolic Diseases
- Subjects
Male ,Proteomics ,Ribosomal Proteins ,0301 basic medicine ,Mitochondrial DNA ,Mitochondrial Diseases ,Adolescent ,Mitochondrial translation ,Protein subunit ,RNA Splicing ,Respiratory chain ,Biology ,Mitochondrion ,Compound heterozygosity ,DNA, Mitochondrial ,Article ,Oxidative Phosphorylation ,Mitochondrial Proteins ,03 medical and health sciences ,0302 clinical medicine ,Ribosomal protein ,Genetics ,Mitochondrial ribosome ,medicine ,Humans ,Exome ,Leigh disease ,Child ,Genetics (clinical) ,Ribosome Subunits, Small, Eukaryotic ,Base Sequence ,Correction ,Infant ,Sequence Analysis, DNA ,medicine.disease ,Molecular biology ,Human genetics ,Mitochondria ,3. Good health ,030104 developmental biology ,Child, Preschool ,Female ,Leigh Disease ,030217 neurology & neurosurgery - Abstract
The synthesis of all 13 mitochondrial DNA (mtDNA)-encoded protein subunits of the human oxidative phosphorylation (OXPHOS) system is carried out by mitochondrial ribosomes (mitoribosomes). Defects in the stability of mitoribosomal proteins or mitoribosome assembly impair mitochondrial protein translation, causing combined OXPHOS enzyme deficiency and clinical disease. Here we report four autosomal-recessive pathogenic mutations in the gene encoding the small mitoribosomal subunit protein, MRPS34, in six subjects from four unrelated families with Leigh syndrome and combined OXPHOS defects. Whole-exome sequencing was used to independently identify all variants. Two splice-site mutations were identified, including homozygous c.321+1G>T in a subject of Italian ancestry and homozygous c.322−10G>A in affected sibling pairs from two unrelated families of Puerto Rican descent. In addition, compound heterozygous MRPS34 mutations were identified in a proband of French ancestry; a missense (c.37G>A [p.Glu13Lys]) and a nonsense (c.94C>T [p.Gln32∗]) variant. We demonstrated that these mutations reduce MRPS34 protein levels and the synthesis of OXPHOS subunits encoded by mtDNA. Examination of the mitoribosome profile and quantitative proteomics showed that the mitochondrial translation defect was caused by destabilization of the small mitoribosomal subunit and impaired monosome assembly. Lentiviral-mediated expression of wild-type MRPS34 rescued the defect in mitochondrial translation observed in skin fibroblasts from affected subjects, confirming the pathogenicity of MRPS34 mutations. Our data establish that MRPS34 is required for normal function of the mitoribosome in humans and furthermore demonstrate the power of quantitative proteomic analysis to identify signatures of defects in specific cellular pathways in fibroblasts from subjects with inherited disease.
- Published
- 2017
18. Defective mitochondrial rRNA methyltransferase MRM2 causes MELAS-like clinical syndrome
- Author
-
Salvatore DiMauro, Martino Montomoli, Michal Minczuk, Cristina Dallabona, Tiziana Lodi, Sarah E. Calvo, Pedro Rebelo-Guiomar, Joanna Rorbach, Ileana Ferrero, Elena Procopio, Massimo Zeviani, Caterina Garone, Maria Alice Donati, Renzo Guerrini, Vamsi K. Mootha, Aaron R. D’Souza, Garone C., D'Souza A.R., Dallabona C., Lodi T., Rebelo-Guiomar P., Rorbach J., Donati M.A., Procopio E., Montomoli M., Guerrini R., Zeviani M., Calvo S.E., Mootha V.K., DiMauro S., Ferrero I., Minczuk M., Garone, Caterina [0000-0003-4928-1037], Minczuk, Michal [0000-0001-8242-1420], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Male ,16S ,Mitochondrial translation ,Saccharomyces cerevisiae ,Mitochondrion ,Biology ,medicine.disease_cause ,MELAS syndrome ,DNA, Mitochondrial ,Mitochondrial Encephalomyopathie ,03 medical and health sciences ,Mitochondrial Encephalomyopathies ,RNA, Ribosomal, 16S ,Genetics ,medicine ,MELAS Syndrome ,Humans ,Amino Acid Sequence ,Child ,Methyltransferase ,Molecular Biology ,Gene ,Genetics (clinical) ,Exome sequencing ,Nuclear Protein ,Ribosomal ,Mutation ,Methyltransferases ,Mitochondria ,Nuclear Proteins ,RNA, Ribosomal ,General Medicine ,DNA ,Articles ,medicine.disease ,Molecular biology ,3. Good health ,Mitochondrial ,Complementation ,030104 developmental biology ,RNA ,Human - Abstract
Defects in nuclear-encoded proteins of the mitochondrial translation machinery cause early-onset and tissue-specific deficiency of one or more OXPHOS complexes. Here, we report a 7-year-old Italian boy with childhood-onset rapidly progressive encepha- lomyopathy and stroke-like episodes. Multiple OXPHOS defects and decreased mtDNA copy number (40%) were detected in muscle homogenate. Clinical features combined with low level of plasma citrulline were highly suggestive of mitochondrial en- cephalopathy, lactic acidosis and stroke-like episodes (MELAS) syndrome, however, the common m.3243 A > G mutation was ex- cluded. Targeted exome sequencing of genes encoding the mitochondrial proteome identified a damaging mutation, c.567 G > A, affecting a highly conserved amino acid residue (p.Gly189Arg) of the MRM2 protein. MRM2 has never before been linked to a hu- man disease and encodes an enzyme responsible for 2’-O-methyl modification at position U1369 in the human mitochondrial 16S rRNA. We generated a knockout yeast model for the orthologous gene that showed a defect in respiration and the reduction of the 2’-O-methyl modification at the equivalent position (U2791) in the yeast mitochondrial 21S rRNA. Complementation with the mrm2 allele carrying the equivalent yeast mutation failed to rescue the respiratory phenotype, which was instead com- pletely rescued by expressing the wild-type allele. Our findings establish that defective MRM2 causes a MELAS-like phenotype, and suggests the genetic screening of the MRM2 gene in patients with a m.3243 A > G negative MELAS-like presentation.
- Published
- 2017
19. Comparative Analysis of Mitochondrial N-Termini from Mouse, Human, and Yeast
- Author
-
Vamsi K. Mootha, Olivier Julien, Karl R. Clauser, Sarah E. Calvo, James A. Wells, Kimberli J. Kamer, and Hongying Shen
- Subjects
Proteomics ,0301 basic medicine ,Saccharomyces cerevisiae Proteins ,Amino Acid Motifs ,Saccharomyces cerevisiae ,Mitochondrion ,Kidney ,Cleavage (embryo) ,Biochemistry ,Cell Line ,Analytical Chemistry ,Conserved sequence ,Evolution, Molecular ,Mitochondrial Proteins ,Mice ,03 medical and health sciences ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Peptide sequence ,Conserved Sequence ,chemistry.chemical_classification ,Chemistry ,Research ,Kidney metabolism ,Mitochondria ,Cell biology ,Amino acid ,030104 developmental biology ,Liver ,Cytoplasm ,OGDH - Abstract
The majority of mitochondrial proteins are encoded in the nuclear genome, translated in the cytoplasm, and directed to the mitochondria by an N-terminal presequence that is cleaved upon import. Recently, N-proteome catalogs have been generated for mitochondria from yeast and from human U937 cells. Here, we applied the subtiligase method to determine N-termini for 327 proteins in mitochondria isolated from mouse liver and kidney. Comparative analysis between mitochondrial N-termini from mouse, human, and yeast proteins shows that whereas presequences are poorly conserved at the sequence level, other presequence properties are extremely conserved, including a length of ∼20-60 amino acids, a net charge between +3 to +6, and the presence of stabilizing amino acids at the N-terminus of mature proteins that follow the N-end rule from bacteria. As in yeast, ∼80% of mouse presequence cleavage sites match canonical motifs for three mitochondrial peptidases (MPP, Icp55, and Oct1), whereas the remainder do not match any known peptidase motifs. We show that mature mitochondrial proteins often exist with a spectrum of N-termini, consistent with a model of multiple cleavage events by MPP and Icp55. In addition to analysis of canonical targeting presequences, our N-terminal dataset allows the exploration of other cleavage events and provides support for polypeptide cleavage into two distinct enzymes (Hsd17b4), protein cleavages key for signaling (Oma1, Opa1, Htra2, Mavs, and Bcs2l13), and in several cases suggests novel protein isoforms (Scp2, Acadm, Adck3, Hsdl2, Dlst, and Ogdh). We present an integrated catalog of mammalian mitochondrial N-termini that can be used as a community resource to investigate individual proteins, to elucidate mechanisms of mammalian mitochondrial processing, and to allow researchers to engineer tags distally to the presequence cleavage.
- Published
- 2017
20. A patient with homozygous nonsense variants in two Leigh syndrome disease genes: Distinguishing a dual diagnosis from a hypomorphic protein-truncating variant
- Author
-
Michael T. Ryan, David A. Stroud, Nicole J. Lake, Vamsi K. Mootha, John Christodoulou, David R. Thorburn, Bharti Morar, Peter Procopis, Luke E. Formosa, Alison G. Compton, and Sarah E. Calvo
- Subjects
medicine.medical_specialty ,Mitochondrial disease ,media_common.quotation_subject ,Nonsense ,Pyruvate Dehydrogenase Complex ,Biology ,medicine.disease_cause ,Mitochondrial Membrane Transport Proteins ,Article ,03 medical and health sciences ,Gene Knockout Techniques ,Mutant protein ,Mitochondrial Precursor Protein Import Complex Proteins ,Exome Sequencing ,Genetics ,medicine ,Humans ,Leigh disease ,Gene ,Genetics (clinical) ,Exome sequencing ,030304 developmental biology ,media_common ,Sequence Deletion ,0303 health sciences ,Mutation ,030305 genetics & heredity ,Homozygote ,medicine.disease ,3. Good health ,Early Diagnosis ,HEK293 Cells ,Medical genetics ,Leigh Disease - Abstract
Leigh syndrome is a mitochondrial disease caused by pathogenic variants in over 85 genes. Whole exome sequencing of a patient with Leigh-like syndrome identified homozygous protein-truncating variants in two genes associated with Leigh syndrome; a reported pathogenic variant in PDHX (NP_003468.2:p.(Arg446*)), and an uncharacterized variant in complex I (CI) assembly factor TIMMDC1 (NP_057673.2:p.(Arg225*)). The TIMMDC1 variant was predicted to truncate 61 amino acids at the C-terminus and functional studies demonstrated a hypomorphic impact of the variant on CI assembly. However, the mutant protein could still rescue CI assembly in TIMMDC1 knockout cells and the patient's clinical phenotype was not clearly distinct from that of other patients with the same PDHX defect. Our data suggest that the hypomorphic effect of the TIMMDC1 protein-truncating variant does not constitute a dual diagnosis in this individual. We recommend cautious assessment of variants in the C-terminus of TIMMDC1 and emphasize the need to consider the caveats detailed within the American College of Medical Genetics and Genomics (ACMG) criteria when assessing variants.
- Published
- 2018
21. Early loss of mitochondrial complex I and rewiring of glutathione metabolism in renal oncocytoma
- Author
-
Vamsi K. Mootha, Sarah E. Calvo, Yang Li, Eliezer M. Van Allen, Eran Mick, Declan McGuone, Kerry A. Pierce, Angela R. Shih, Esther Oliva, Clary B. Clish, Frances L Chaves, Raj K. Gopal, and Andrea Garofalo
- Subjects
Male ,0301 basic medicine ,Cell Survival ,Nephron ,Mitochondrion ,Biology ,DNA, Mitochondrial ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,medicine ,Metabolome ,Adenoma, Oxyphilic ,Humans ,Cyclin D1 ,Oncocytoma ,Renal oncocytoma ,Electron Transport Complex I ,Multidisciplinary ,Gene Expression Profiling ,DNA, Neoplasm ,Glutathione ,medicine.disease ,Molecular biology ,Kidney Neoplasms ,Mitochondria ,Neoplasm Proteins ,030104 developmental biology ,medicine.anatomical_structure ,GCLC ,chemistry ,Chromosomes, Human, Pair 1 ,Female - Abstract
Renal oncocytomas are benign tumors characterized by a marked accumulation of mitochondria. We report a combined exome, transcriptome, and metabolome analysis of these tumors. Joint analysis of the nuclear and mitochondrial (mtDNA) genomes reveals loss-of-function mtDNA mutations occurring at high variant allele fractions, consistent with positive selection, in genes encoding complex I as the most frequent genetic events. A subset of these tumors also exhibits chromosome 1 loss and/or cyclin D1 overexpression, suggesting they follow complex I loss. Transcriptome data revealed that many pathways previously reported to be altered in renal oncocytoma were simply differentially expressed in the tumor's cell of origin, the distal nephron, compared with other nephron segments. Using a heuristic approach to account for cell-of-origin bias we uncovered strong expression alterations in the gamma-glutamyl cycle, including glutathione synthesis (increased GCLC) and glutathione degradation. Moreover, the most striking changes in metabolite profiling were elevations in oxidized and reduced glutathione as well as γ-glutamyl-cysteine and cysteinyl-glycine, dipeptide intermediates in glutathione biosynthesis, and recycling, respectively. Biosynthesis of glutathione appears adaptive as blockade of GCLC impairs viability in cells cultured with a complex I inhibitor. Our data suggest that loss-of-function mutations in complex I are a candidate driver event in renal oncocytoma that is followed by frequent loss of chromosome 1, cyclin D1 overexpression, and adaptive up-regulation of glutathione biosynthesis.
- Published
- 2018
22. GeNets: a unified web platform for network-based genomic analyses
- Author
-
Joseph Rosenbluh, Ted Liefeld, Taibo Li, Ayshwarya Subramanian, David An, Arthur Liberzon, Heiko Horn, Aviv Regev, Dawn A. Thompson, Kasper Lage, Jon Bistline, Aviad Tsherniak, Rajiv Narayan, Jesse S. Boehm, Nir Hacohen, Liraz Greenfeld, Jacob D. Jaffe, Andrew Zimmer, April Kim, Sarah E. Calvo, Jill P. Mesirov, Bang Wong, Yang Li, Steve Carr, Ted Natoli, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Li, Taibo, and Regev, Aviv
- Subjects
0301 basic medicine ,business.industry ,Extramural ,Computer science ,Systems biology ,Cell Biology ,Nucleic acid amplification technique ,Computational biology ,Network topology ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Software ,ComputingMethodologies_PATTERNRECOGNITION ,Dna genetics ,The Internet ,business ,Molecular Biology ,Functional genomics ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Functional genomics networks are widely used to identify unexpected pathway relationships in large genomic datasets. However, it is challenging to compare the signal-to-noise ratios of different networks and to identify the optimal network with which to interpret a particular genetic dataset. We present GeNets, a platform in which users can train a machine-learning model (Quack) to carry out these comparisons and execute, store, and share analyses of genetic and RNA-sequencing datasets.
- Published
- 2018
23. GeNets: A unified web platform for network-based analyses of genomic data
- Author
-
Jill P. Mesirov, Kasper Lage, Ted Natoli, April Kim, Rajiv Narayan, Liraz Greenfeld, Nir Hacohen, Jacob D. Jaffe, Arthur Liberzon, Jon Bistline, Ted Liefeld, Aviad Tsherniak, Heiko Horn, Sarah E. Calvo, Yang Li, Steve Carr, Bang Wong, Andrew Zimmer, Ayshwarya Subramanian, Taibo Li, Dawn A. Thompson, Jesse S. Boehm, Aviv Regev, Joseph Rosenbluh, and David An
- Subjects
Genomic data ,Genomics ,Biology ,computer.software_genre ,Article ,Bottleneck ,Machine Learning ,03 medical and health sciences ,0302 clinical medicine ,030304 developmental biology ,Internet ,0303 health sciences ,SIGNAL (programming language) ,DNA ,Pathway information ,ComputingMethodologies_PATTERNRECOGNITION ,Workflow ,Scalability ,RNA ,Data mining ,Databases, Nucleic Acid ,Nucleic Acid Amplification Techniques ,Functional genomics ,computer ,Software ,030217 neurology & neurosurgery - Abstract
A major bottleneck in network-based analyses of genomic data is quantitatively comparing biological signal in different networks and to identifying the optimal network dataset to answer a particular biological question. Towards these aims, we developed a unified web platform 9Broad Institute Web Platform for Genome Networks (GeNets)9, where users can compare biological signal of networks, and execute, store, and share network analyses. We designed a machine learningmachine-learning algorithm (Quack) which), which uses topological features to can quantify the overall and pathway-specific biological signals in networks, thus enabling users to choose the optimal network dataset for their analyses. We illustrated a typical workflow using GeNets to identify interesting autism candidate genes in the network that, when compared to four other networks, best recapitulates established neurodevelopmental pathway information. GeNets is a scalable, general and uniquely enabling computational framework for analyzing, managing and sharing analyses of genetic datasets using heterogeneous functional genomics networks, for example, from single-cell transcriptional analyses.
- Published
- 2018
24. Bayesian Hidden Markov Tree Models for Clustering Genes with Shared Evolutionary History
- Author
-
Sarah E. Calvo, Jun Liu, Shaoyang Ning, Yang Li, and Vamsi K. Mootha
- Subjects
0301 basic medicine ,Statistics and Probability ,FOS: Computer and information sciences ,Computer science ,Bayesian probability ,Bayesian inference ,Machine learning ,computer.software_genre ,Statistics - Applications ,01 natural sciences ,010104 statistics & probability ,03 medical and health sciences ,symbols.namesake ,Applications (stat.AP) ,0101 mathematics ,Cluster analysis ,Hidden Markov model ,gene function prediction ,business.industry ,Statistical model ,Dirichlet process ,Co-evolution ,tree-structured hidden Markov model ,030104 developmental biology ,Dirichlet process mixture model ,Modeling and Simulation ,symbols ,Artificial intelligence ,Statistics, Probability and Uncertainty ,business ,evolutionary history ,computer ,Gibbs sampling ,Clime - Abstract
Determination of functions for poorly characterized genes is crucial for understanding biological processes and studying human diseases. Functionally associated genes are often gained and lost together through evolution. Therefore identifying co-evolution of genes can predict functional gene-gene associations. We describe here the full statistical model and computational strategies underlying the original algorithm, CLustering by Inferred Models of Evolution (CLIME 1.0) recently reported by us [Li et al., 2014]. CLIME 1.0 employs a mixture of tree-structured hidden Markov models for gene evolution process, and a Bayesian model-based clustering algorithm to detect gene modules with shared evolutionary histories (termed evolutionary conserved modules, or ECMs). A Dirichlet process prior was adopted for estimating the number of gene clusters and a Gibbs sampler was developed for posterior sampling. We further developed an extended version, CLIME 1.1, to incorporate the uncertainty on the evolutionary tree structure. By simulation studies and benchmarks on real data sets, we show that CLIME 1.0 and CLIME 1.1 outperform traditional methods that use simple metrics (e.g., the Hamming distance or Pearson correlation) to measure co-evolution between pairs of genes., Comment: 34 pages, 8 figures
- Published
- 2018
- Full Text
- View/download PDF
25. Widespread Chromosomal Losses and Mitochondrial DNA Alterations as Genetic Drivers in Hürthle Cell Carcinoma
- Author
-
Gilbert H. Daniels, Julian M. Hess, Raj K. Gopal, Salma Amin, Peter F. Arndt, Sarah E. Calvo, Carrie C. Lubitz, Jiwoong Kim, Dora Dias-Santagata, Dimitri Livitz, David G. McFadden, Lori J. Wirth, Braidie Campbell, Sareh Parangi, Paz Polak, Lior Z. Braunstein, A. John Iafrate, Scott Mordecai, Vamsi K. Mootha, Benjamin J. Gigliotti, Ignaty Leshchiner, Angela R. Shih, Tiannan Zhan, Frances L Chaves, Daniel Rosebrock, Zenon Grabarek, K Kübler, Gad Getz, Samuel E. Donovan, Chip Stewart, and Peter M. Sadow
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,Mitochondrial DNA ,DNA Copy Number Variations ,Mitochondrion ,Biology ,Haploidy ,DNA, Mitochondrial ,Article ,Metastasis ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Death-associated protein 6 ,Exome Sequencing ,medicine ,Humans ,Thyroid Neoplasms ,Neoplasm Metastasis ,Thyroid cancer ,Telomerase ,Chromosome Aberrations ,Thyroid ,Cell Biology ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research - Abstract
Summary Hurthle cell carcinoma of the thyroid (HCC) is a form of thyroid cancer recalcitrant to radioiodine therapy that exhibits an accumulation of mitochondria. We performed whole-exome sequencing on a cohort of primary, recurrent, and metastatic tumors, and identified recurrent mutations in DAXX, TP53, NRAS, NF1, CDKN1A, ARHGAP35, and the TERT promoter. Parallel analysis of mtDNA revealed recurrent homoplasmic mutations in subunits of complex I of the electron transport chain. Analysis of DNA copy-number alterations uncovered widespread loss of chromosomes culminating in near-haploid chromosomal content in a large fraction of HCC, which was maintained during metastatic spread. This work uncovers a distinct molecular origin of HCC compared with other thyroid malignancies.
- Published
- 2017
26. MitoCarta2.0: an updated inventory of mammalian mitochondrial proteins
- Author
-
Karl R. Clauser, Vamsi K. Mootha, and Sarah E. Calvo
- Subjects
0301 basic medicine ,Genetics ,Internet ,Bayes Theorem ,Genomics ,Mitochondrion ,Biology ,Homology (biology) ,Mitochondrial Proteins ,Mice ,03 medical and health sciences ,030104 developmental biology ,Ion homeostasis ,Organelle ,Database Issue ,Animals ,Humans ,Human genome ,Signal transduction ,Databases, Protein ,Gene - Abstract
Mitochondria are complex organelles that house essential pathways involved in energy metabolism, ion homeostasis, signalling and apoptosis. To understand mitochondrial pathways in health and disease, it is crucial to have an accurate inventory of the organelle's protein components. In 2008, we made substantial progress toward this goal by performing in-depth mass spectrometry of mitochondria from 14 organs, epitope tagging/microscopy and Bayesian integration to assemble MitoCarta (www.broadinstitute.org/pubs/MitoCarta): an inventory of genes encoding mitochondrial-localized proteins and their expression across 14 mouse tissues. Using the same strategy we have now reconstructed this inventory separately for human and for mouse based on (i) improved gene transcript models, (ii) updated literature curation, including results from proteomic analyses of mitochondrial sub-compartments, (iii) improved homology mapping and (iv) updated versions of all seven original data sets. The updated human MitoCarta2.0 consists of 1158 human genes, including 918 genes in the original inventory as well as 240 additional genes. The updated mouse MitoCarta2.0 consists of 1158 genes, including 967 genes in the original inventory plus 191 additional genes. The improved MitoCarta 2.0 inventory provides a molecular framework for system-level analysis of mammalian mitochondria.
- Published
- 2015
27. CLIC, a tool for expanding biological pathways based on co-expression across thousands of datasets
- Author
-
Alexis A. Jourdain, Sarah E. Calvo, Vamsi K. Mootha, Jun Liu, and Yang Li
- Subjects
Metabolic Processes ,0301 basic medicine ,Databases, Factual ,Microarrays ,Normal Distribution ,Gene regulatory network ,Gene Expression ,Biochemistry ,Oxidative Phosphorylation ,0302 clinical medicine ,Cluster Analysis ,Gene Regulatory Networks ,lcsh:QH301-705.5 ,Energy-Producing Organelles ,Genetics ,Ecology ,Genomics ,Mitochondria ,Precipitation Techniques ,Bioassays and Physiological Analysis ,Computational Theory and Mathematics ,Modeling and Simulation ,Physical Sciences ,Cellular Structures and Organelles ,DNA microarray ,Algorithms ,Research Article ,Signal Transduction ,Gene prediction ,Posterior probability ,Bayesian probability ,Computational biology ,Bioenergetics ,Biology ,Research and Analysis Methods ,Models, Biological ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Immunoprecipitation ,Humans ,Gene Regulation ,Gene Prediction ,Cluster analysis ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Gene Expression Profiling ,Biology and Life Sciences ,Computational Biology ,Proteins ,Protein Complexes ,Proteasomes ,Cell Biology ,Genome Analysis ,Probability Theory ,Probability Distribution ,Co-Immunoprecipitation ,Gene expression profiling ,Metabolism ,030104 developmental biology ,lcsh:Biology (General) ,Transcriptome ,Mathematics ,Software ,030217 neurology & neurosurgery - Abstract
In recent years, there has been a huge rise in the number of publicly available transcriptional profiling datasets. These massive compendia comprise billions of measurements and provide a special opportunity to predict the function of unstudied genes based on co-expression to well-studied pathways. Such analyses can be very challenging, however, since biological pathways are modular and may exhibit co-expression only in specific contexts. To overcome these challenges we introduce CLIC, CLustering by Inferred Co-expression. CLIC accepts as input a pathway consisting of two or more genes. It then uses a Bayesian partition model to simultaneously partition the input gene set into coherent co-expressed modules (CEMs), while assigning the posterior probability for each dataset in support of each CEM. CLIC then expands each CEM by scanning the transcriptome for additional co-expressed genes, quantified by an integrated log-likelihood ratio (LLR) score weighted for each dataset. As a byproduct, CLIC automatically learns the conditions (datasets) within which a CEM is operative. We implemented CLIC using a compendium of 1774 mouse microarray datasets (28628 microarrays) or 1887 human microarray datasets (45158 microarrays). CLIC analysis reveals that of 910 canonical biological pathways, 30% consist of strongly co-expressed gene modules for which new members are predicted. For example, CLIC predicts a functional connection between protein C7orf55 (FMC1) and the mitochondrial ATP synthase complex that we have experimentally validated. CLIC is freely available at www.gene-clic.org. We anticipate that CLIC will be valuable both for revealing new components of biological pathways as well as the conditions in which they are active., Author summary A major challenge in modern genomics research is to link the thousands of unstudied genes to the pathways and complexes within which they operate. A popular strategy to infer the function of an unstudied gene is to search for co-expressing genes of known function using a single transcriptional profiling dataset. Today, there are literally thousands of transcriptional profiling datasets, and a special opportunity lies in querying entire compendia for co-expression in order to more reliably expand pathway membership. Such analyses can be challenging, however, as pathways can be highly modular, and different datasets can conflict in terms of providing evidence of co-expression. To overcome these challenges, we introduce a tool called CLIC, CLustering by Inferred Co-expression. CLIC accepts a pathway of interest, simultaneously partitioning it into modules of genes that exhibit striking co-expression patterns while also learning the number of modules. It then expands each module with new members, based on an integrated weighted co-expression score across the datasets. Three key innovations within CLIC–partitioning, background correction, and integration–distinguish it from other methods. A side benefit of CLIC is that it spotlights the datasets that support the co-expression of a given co-expression module. Our software is freely available, and should be useful for identifying new genes in biological pathways while also identifying the datasets within which the pathways are active.
- Published
- 2017
28. Mutation in the novel nuclear-encoded mitochondrial protein CHCHD10 in a family with autosomal dominant mitochondrial myopathy
- Author
-
Teepu Siddique, Albert J. Tahmoush, Faisal Fecto, Irfan Lalani, Han Xiang Deng, Kaouther Ajroud, Vamsi K. Mootha, Sarah E. Calvo, Senda Ajroud-Driss, Terry Heiman-Patterson, and Nailah Siddique
- Subjects
Male ,Candidate gene ,Nuclear gene ,Chromosomes, Human, Pair 22 ,Mitochondrial disease ,Mutant ,Locus (genetics) ,Mitochondrion ,Biology ,Human mitochondrial genetics ,Article ,Mitochondrial Proteins ,Cellular and Molecular Neuroscience ,Mitochondrial myopathy ,Genetics ,medicine ,Humans ,Family ,Genetics (clinical) ,Genes, Dominant ,Puerto Rico ,Mitochondrial Myopathies ,medicine.disease ,Molecular biology ,Mitochondria ,Mutation ,Female - Abstract
Mitochondrial myopathies belong to a larger group of systemic diseases caused by morphological or biochemical abnormalities of mitochondria. Mitochondrial disorders can be caused by mutations in either the mitochondrial or nuclear genome. Only 5 % of all mitochondrial disorders are autosomal dominant. We analyzed DNA from members of the previously reported Puerto Rican kindred with an autosomal dominant mitochondrial myopathy (Heimann-Patterson et al. 1997). Linkage analysis suggested a putative locus on the pericentric region of the long arm of chromosome 22 (22q11). Using the tools of integrative genomics, we established chromosome 22 open reading frame 16 (C22orf16) (later designated as CHCHD10) as the only high-scoring mitochondrial candidate gene in our minimal candidate region. Sequence analysis revealed a double-missense mutation (R15S and G58R) in cis in CHCHD10 which encodes a coiled coil-helix-coiled coil-helix protein of unknown function. These two mutations completely co-segregated with the disease phenotype and were absent in 1,481 Caucasian and 80 Hispanic (including 32 Puerto Rican) controls. Expression profiling showed that CHCHD10 is enriched in skeletal muscle. Mitochondrial localization of the CHCHD10 protein was confirmed using immunofluorescence in cells expressing either wild-type or mutant CHCHD10. We found that the expression of the G58R, but not the R15S, mutation induced mitochondrial fragmentation. Our findings identify a novel gene causing mitochondrial myopathy, thereby expanding the spectrum of mitochondrial myopathies caused by nuclear genes. Our findings also suggest a role for CHCHD10 in the morphologic remodeling of the mitochondria.
- Published
- 2014
29. Expansion of Biological Pathways Based on Evolutionary Inference
- Author
-
Yang Li, Jun Liu, Vamsi K. Mootha, Sarah E. Calvo, and Roee Gutman
- Subjects
Proteome ,Plasmodium falciparum ,Inference ,Computational algorithm ,Computational biology ,Biology ,Shared history ,Genome ,General Biochemistry, Genetics and Molecular Biology ,Homology (biology) ,03 medical and health sciences ,0302 clinical medicine ,Phylogenetics ,Yeasts ,Cluster Analysis ,Humans ,Cluster analysis ,Phylogeny ,030304 developmental biology ,Genetics ,0303 health sciences ,Biochemistry, Genetics and Molecular Biology(all) ,Mitochondria ,3. Good health ,Rhodophyta ,Algorithms ,030217 neurology & neurosurgery ,Signal Transduction ,Clime - Abstract
SummaryThe availability of diverse genomes makes it possible to predict gene function based on shared evolutionary history. This approach can be challenging, however, for pathways whose components do not exhibit a shared history but rather consist of distinct “evolutionary modules.” We introduce a computational algorithm, clustering by inferred models of evolution (CLIME), which inputs a eukaryotic species tree, homology matrix, and pathway (gene set) of interest. CLIME partitions the gene set into disjoint evolutionary modules, simultaneously learning the number of modules and a tree-based evolutionary history that defines each module. CLIME then expands each module by scanning the genome for new components that likely arose under the inferred evolutionary model. Application of CLIME to ∼1,000 annotated human pathways and to the proteomes of yeast, red algae, and malaria reveals unanticipated evolutionary modularity and coevolving components. CLIME is freely available and should become increasingly powerful with the growing wealth of eukaryotic genomes.
- Published
- 2014
30. Hypoxia Rescues Frataxin Loss by Restoring Iron Sulfur Cluster Biogenesis
- Author
-
Vamsi K. Mootha, David P. Barondeau, Hong Wang, Tslil Ast, Robert M. H. Grange, Joshua D. Meisel, Sarah E. Calvo, Gary Ruvkun, Sharon H. Kim, Fumito Ichinose, Fumiaki Nagashima, Lauren L. Orefice, Shachin Patra, and Warren M. Zapol
- Subjects
Iron-Sulfur Proteins ,Male ,Ataxia ,NF-E2-Related Factor 2 ,Iron ,Iron–sulfur cluster ,Saccharomyces cerevisiae ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mitochondrial Proteins ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Iron-Binding Proteins ,medicine ,Animals ,Humans ,Caenorhabditis elegans ,Hypoxia ,Iron Regulatory Protein 2 ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,biology ,ATF4 ,HEK 293 cells ,Activating Transcription Factor 4 ,Mitochondria ,Cell biology ,Oxidative Stress ,HEK293 Cells ,chemistry ,Friedreich Ataxia ,Frataxin ,biology.protein ,Female ,medicine.symptom ,K562 Cells ,Sulfur ,030217 neurology & neurosurgery ,Biogenesis ,Oxidative stress ,K562 cells - Abstract
Summary Friedreich’s ataxia (FRDA) is a devastating, multisystemic disorder caused by recessive mutations in the mitochondrial protein frataxin (FXN). FXN participates in the biosynthesis of Fe-S clusters and is considered to be essential for viability. Here we report that when grown in 1% ambient O2, FXN null yeast, human cells, and nematodes are fully viable. In human cells, hypoxia restores steady-state levels of Fe-S clusters and normalizes ATF4, NRF2, and IRP2 signaling events associated with FRDA. Cellular studies and in vitro reconstitution indicate that hypoxia acts through HIF-independent mechanisms that increase bioavailable iron as well as directly activate Fe-S synthesis. In a mouse model of FRDA, breathing 11% O2 attenuates the progression of ataxia, whereas breathing 55% O2 hastens it. Our work identifies oxygen as a key environmental variable in the pathogenesis associated with FXN depletion, with important mechanistic and therapeutic implications.
- Published
- 2019
31. EMRE Is an Essential Component of the Mitochondrial Calcium Uniporter Complex
- Author
-
Yasemin Sancak, Namrata D. Udeshi, Sarah E. Calvo, Toshimori Kitami, Andrew L. Markhard, Olga Goldberger, Steven A. Carr, David E. Clapham, Andrew A. Li, Kimberli J. Kamer, Erika Kovács-Bogdán, Dipayan Chaudhuri, and Vamsi K. Mootha
- Subjects
Proteomics ,Molecular Sequence Data ,Mitochondrion ,LETM1 ,Mitochondrial Membrane Transport Proteins ,Article ,Mitochondrial membrane transport protein ,Calcium-binding protein ,Humans ,Inner membrane ,Mitochondrial calcium uptake ,Amino Acid Sequence ,EF Hand Motifs ,Uniporter ,Cation Transport Proteins ,Phylogeny ,Multidisciplinary ,biology ,Calcium channel ,Calcium-Binding Proteins ,Cell Membrane ,Mitochondria ,Protein Structure, Tertiary ,Cell biology ,HEK293 Cells ,Gene Knockdown Techniques ,biology.protein ,Calcium Channels - Abstract
The mitochondrial uniporter is a highly selective calcium channel in the organelle's inner membrane. Its molecular components include the EF-hand-containing calcium-binding proteins mitochondrial calcium uptake 1 (MICU1) and MICU2 and the pore-forming subunit mitochondrial calcium uniporter (MCU). We sought to achieve a full molecular characterization of the uniporter holocomplex (uniplex). Quantitative mass spectrometry of affinity-purified uniplex recovered MICU1 and MICU2, MCU and its paralog MCUb, and essential MCU regulator (EMRE), a previously uncharacterized protein. EMRE is a 10-kilodalton, metazoan-specific protein with a single transmembrane domain. In its absence, uniporter channel activity was lost despite intact MCU expression and oligomerization. EMRE was required for the interaction of MCU with MICU1 and MICU2. Hence, EMRE is essential for in vivo uniporter current and additionally bridges the calcium-sensing role of MICU1 and MICU2 with the calcium-conducting role of MCU.
- Published
- 2013
32. A Genome-wide CRISPR Death Screen Identifies Genes Essential for Oxidative Phosphorylation
- Author
-
Vamsi K. Mootha, David E. Root, John G. Doench, Jason D. Arroyo, Sarah E. Calvo, Alexis A. Jourdain, and Carmine A. Ballarano
- Subjects
0301 basic medicine ,Physiology ,Synthetic lethality ,Mitochondrion ,Biology ,Genome ,Article ,Oxidative Phosphorylation ,03 medical and health sciences ,RNA, Ribosomal, 16S ,Mitochondrial ribosome ,CRISPR ,Humans ,Clustered Regularly Interspaced Short Palindromic Repeats ,Molecular Biology ,Gene ,Genetics ,Cell Death ,Galactose ,Reproducibility of Results ,Translation (biology) ,Cell Biology ,Mitochondria ,030104 developmental biology ,Genes, Mitochondrial ,Glucose ,HEK293 Cells ,Phenotype ,Protein Biosynthesis ,Human genome ,K562 Cells ,HeLa Cells - Abstract
Summary Oxidative phosphorylation (OXPHOS) is the major pathway for ATP production in humans. Deficiencies in OXPHOS can arise from mutations in either mitochondrial or nuclear genomes and comprise the largest collection of inborn errors of metabolism. At present we lack a complete catalog of human genes and pathways essential for OXPHOS. Here we introduce a genome-wide CRISPR "death screen" that actively selects dying cells to reveal human genes required for OXPHOS, inspired by the classic observation that human cells deficient in OXPHOS survive in glucose but die in galactose. We report 191 high-confidence hits essential for OXPHOS, including 72 underlying known OXPHOS diseases. Our screen reveals a functional module consisting of NGRN, WBSCR16, RPUSD3, RPUSD4, TRUB2, and FASTKD2 that regulates the mitochondrial 16S rRNA and intra-mitochondrial translation. Our work yields a rich catalog of genes required for OXPHOS and, more generally, demonstrates the power of death screening for functional genomic analysis.
- Published
- 2016
33. Comparative RNA editing in autistic and neurotypical cerebella
- Author
-
Kayla Vatalaro, Fedik Rahimov, Emery N. Brown, Jillian McCarthy, David M. Margulies, Sarah E. Calvo, Kyriacos Markianos, Alal Eran, Jin Billy Li, Louis M. Kunkel, Isaac S. Kohane, Christin D. Collins, Harvard University--MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, and Brown, Emery N.
- Subjects
Male ,RNA editing ,Adenosine Deaminase ,DNA Mutational Analysis ,Transcriptome ,0302 clinical medicine ,Neurodevelopmental disorder ,Cerebellum ,Receptor, Serotonin, 5-HT2C ,Protein Isoforms ,Child ,Genetics ,0303 health sciences ,education.field_of_study ,neurodevelopment ,RNA-Binding Proteins ,3. Good health ,Psychiatry and Mental health ,Child, Preschool ,Female ,A-to-I ,Neurotypical ,Adolescent ,Filamins ,Population ,autism ,Biology ,Article ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,human cerebellum ,medicine ,Humans ,Receptors, AMPA ,Autistic Disorder ,education ,Molecular Biology ,Gene ,Gene Library ,030304 developmental biology ,epigenetics ,RNA ,Numerical Analysis, Computer-Assisted ,medicine.disease ,Autism ,Kv1.1 Potassium Channel ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Adenosine-to-inosine (A-to-I) RNA editing is a neurodevelopmentally regulated epigenetic modification shown to modulate complex behavior in animals. Little is known about human A-to-I editing, but it is thought to constitute one of many molecular mechanisms connecting environmental stimuli and behavioral outputs. Thus, comprehensive exploration of A-to-I RNA editing in human brains may shed light on gene–environment interactions underlying complex behavior in health and disease. Synaptic function is a main target of A-to-I editing, which can selectively recode key amino acids in synaptic genes, directly altering synaptic strength and duration in response to environmental signals. Here, we performed a high-resolution survey of synaptic A-to-I RNA editing in a human population, and examined how it varies in autism, a neurodevelopmental disorder in which synaptic abnormalities are a common finding. Using ultra-deep (>1000 × ) sequencing, we quantified the levels of A-to-I editing of 10 synaptic genes in postmortem cerebella from 14 neurotypical and 11 autistic individuals. A high dynamic range of editing levels was detected across individuals and editing sites, from 99.6% to below detection limits. In most sites, the extreme ends of the population editing distributions were individuals with autism. Editing was correlated with isoform usage, clusters of correlated sites were identified, and differential editing patterns examined. Finally, a dysfunctional form of the editing enzyme adenosine deaminase acting on RNA B1 was found more commonly in postmortem cerebella from individuals with autism. These results provide a population-level, high-resolution view of A-to-I RNA editing in human cerebella and suggest that A-to-I editing of synaptic genes may be informative for assessing the epigenetic risk for autism., Nancy Lurie Marks Family Foundation, F. Hoffmann-La Roche & Co. (Applied Science Sequencing Grant Program), Autism Speaks (Organization), Simons Foundation, National Institutes of Health (U.S.) (Grant 1R01MH085143-01)
- Published
- 2012
34. Mutations in MTFMT Underlie a Human Disorder of Formylation Causing Impaired Mitochondrial Translation
- Author
-
Jinal Patel, David R. Thorburn, Steven A. Carr, Alison G. Compton, Caroline Köhrer, Matthew McKenzie, Jacob D. Jaffe, Sarah E. Calvo, Jonathon M. Silberstein, Uttam L. RajBhandary, Olga Goldberger, Steven G. Hershman, Vamsi K. Mootha, Michael T. Ryan, John Christodoulou, Elena J. Tucker, and Casey A. Belcher-Timme
- Subjects
Hydroxymethyl and Formyl Transferases ,Mitochondrial DNA ,Heterozygote ,RNA, Transfer, Met ,Mitochondrial translation ,Physiology ,Immunoblotting ,Mitochondrion ,Biology ,DNA, Mitochondrial ,Article ,Mitochondrial Proteins ,03 medical and health sciences ,0302 clinical medicine ,Eukaryotic translation ,Transduction, Genetic ,medicine ,Protein biosynthesis ,Humans ,Leigh disease ,Child ,Molecular Biology ,Cells, Cultured ,030304 developmental biology ,Genetics ,0303 health sciences ,Prokaryotic initiation factor-2 ,Lentivirus ,Virion ,Sequence Analysis, DNA ,Cell Biology ,Fibroblasts ,medicine.disease ,Mitochondria ,Protein Biosynthesis ,Transfer RNA ,Mutation ,Cyclooxygenase 1 ,Leigh Disease ,030217 neurology & neurosurgery - Abstract
The metazoan mitochondrial translation machinery is unusual in having a single tRNA(Met) that fulfills the dual role of the initiator and elongator tRNA(Met). A portion of the Met-tRNA(Met) pool is formylated by mitochondrial methionyl-tRNA formyltransferase (MTFMT) to generate N-formylmethionine-tRNA(Met) (fMet-tRNA(met)), which is used for translation initiation; however, the requirement of formylation for initiation in human mitochondria is still under debate. Using targeted sequencing of the mtDNA and nuclear exons encoding the mitochondrial proteome (MitoExome), we identified compound heterozygous mutations in MTFMT in two unrelated children presenting with Leigh syndrome and combined OXPHOS deficiency. Patient fibroblasts exhibit severe defects in mitochondrial translation that can be rescued by exogenous expression of MTFMT. Furthermore, patient fibroblasts have dramatically reduced fMet-tRNA(Met) levels and an abnormal formylation profile of mitochondrially translated COX1. Our findings demonstrate that MTFMT is critical for efficient human mitochondrial translation and reveal a human disorder of Met-tRNA(Met) formylation.
- Published
- 2011
- Full Text
- View/download PDF
35. The Mitochondrial Proteome and Human Disease
- Author
-
Vamsi K. Mootha and Sarah E. Calvo
- Subjects
Cell Nucleus ,Genetics ,Mitochondrial DNA ,Mitochondrial Diseases ,Proteome ,Mitochondrial disease ,Respiratory chain ,Genomics ,Computational biology ,Biology ,medicine.disease ,Human mitochondrial genetics ,Genome ,Article ,Mitochondria ,Mitochondrial Proteins ,mitochondrial fusion ,Genome, Mitochondrial ,medicine ,Humans ,Molecular Biology ,Genetics (clinical) - Abstract
For nearly three decades, the sequence of the human mitochondrial genome (mtDNA) has provided a molecular framework for understanding maternally inherited diseases. However, the vast majority of human mitochondrial disorders are caused by nuclear genome defects, which is not surprising since the mtDNA encodes only 13 proteins. Advances in genomics, mass spectrometry, and computation have only recently made it possible to systematically identify the complement of over 1,000 proteins that comprise the mammalian mitochondrial proteome. Here, we review recent progress in characterizing the mitochondrial proteome and highlight insights into its complexity, tissue heterogeneity, evolutionary origins, and biochemical versatility. We then discuss how this proteome is being used to discover the genetic basis of respiratory chain disorders as well as to expand our definition of mitochondrial disease. Finally, we explore future prospects and challenges for using the mitochondrial proteome as a foundation for systems analysis of the organelle.
- Published
- 2010
36. Mitochondrial Disease Sequence Data Resource (MSeqDR): a global grass-roots consortium to facilitate deposition, curation, annotation, and integrated analysis of genomic data for the mitochondrial disease clinical and research communities
- Author
-
Xiaowu Gai, Jeremy Leipzig, Aurora Pujol, Richard G. Boles, Deanna M. Church, Finley Macrae, Erin Rooney Riggs, Michio Hirano, Mariangela Santorsola, Yaffa R. Rubinstein, Rosanna Clima, Marie T. Lott, Penelope E. Bonnen, Christine M. Stanley, David Dimmock, Heidi L. Rehm, Marni J. Falk, Giuseppe Gasparre, Holger Prokisch, Shamima Rahman, Claire A. Sheldon, Anu Suomalainen, Hakon Hakonarson, Sarah E. Calvo, Melissa A. Parisi, Alphons P. M. Stassen, Zhe Zhang, Katrina Gwinn, Johan L.K. Van Hove, Lee-Jun C. Wong, Lisa D. Brooks, Virginia Brilhante, William C. Copeland, Jeana T. DaRe, Curt Scharfe, Michael A. Gonzalez, Johan T. den Dunnen, I.F.M. de Coo, Iris L. Gonzalez, Claudia Calabrese, Yasushi Okazaki, Vamsi K. Mootha, Lynne A. Wolfe, Douglas S. Kerr, Doron M. Behar, Bert Smeets, John Christodoulou, Juan C. Perin, Stephan Züchner, Lishuang Shen, Eric A. Shoubridge, Sihoun Hahn, Danuta Krotoski, Sharon F. Terry, Domenico Simone, David Ralph, Renkui Bai, Olga Derbenevoa, Honey V. Reddi, Eric A. Pierce, Daniel Navarro-Gomez, Gregory M. Enns, Vincent Procaccio, Russell P. Saneto, Mannis van Oven, Philip E. Yeske, David R. Thorburn, Bruce H. Cohen, Maria Lvova, Robert Shelton, Douglas C. Wallace, Maria Angela Diroma, Marcella Attimonelli, Dong Li, Elizabeth M. McCormick, Amy Goldstein, Mark A. Tarnopolsky, Patrick F. Chinnery, Donna Maglott, Richard J. Rodenburg, Jirair K. Bedoyan, Richard G.H. Cotton, Richard H. Haas, Jan A.M. Smeitink, Grant A. Mitchell, Isabelle Thiffault, Sherri J. Bale, RS: CARIM - R2 - Cardiac function and failure, RS: GROW - Developmental Biology, Genetica & Celbiologie, Klinische Genetica, RS: GROW - R4 - Reproductive and Perinatal Medicine, Falk M.J., Shen L., Gonzalez M., Leipzig J., Lott M.T., Stassen A.P.M., Diroma M.A., Navarro-Gomez D., Yeske P., Bai R., Boles R.G., Brilhante V., Ralph D., DaRe J.T., Shelton R., Terry S.F., Zhang Z., Copeland W.C., van Oven M., Prokisch H., Wallace D.C., Attimonelli M., Krotoski D., Zuchner S., Gai X., Bale S., Bedoyan J., Behar D., Bonnen P., Brooks L., Calabrese C., Calvo S., Chinnery P., Christodoulou J., Church D.t, Clima R., Cohen B.H., Cotton R.G., de Coo I.F.M., Derbenevoa O., den Dunnen J.T., Dimmock D., Enns G., Gasparre G., Goldstein A., Gonzalez I., Gwinn K., Hahn S., Haas R.H., Hakonarson H., Hirano M., Kerr D., Li D., Lvova M., Macrae F., Maglott D., McCormick E., Mitchell G., Mootha V.K., Okazaki Y., Pujol A., Parisi M., Perin J.C., Pierce E.A., Procaccio V., Rahman S., Reddi H., Rehm H., Riggs E., Rodenburg R., Rubinstein Y., Saneto R., Santorsola M., Scharfe C., Sheldon C., Shoubridge E.A., Simone D., Smeets B., Smeitink J.A., Stanley C., Suomalainen A., Tarnopolsky M., Thiffault I., Thorburn D.R., Hove J.V., Wolfe L., Wong L.J., and Genetic Identification
- Subjects
Male ,Mitochondrial DNA ,Mitochondrial Diseases ,DATABASE ,Endocrinology, Diabetes and Metabolism ,Mitochondrial disease ,Genomics ,mitochondrial DNA ,Computational biology ,Biology ,Biochemistry ,Genome ,Article ,03 medical and health sciences ,Annotation ,User-Computer Interface ,0302 clinical medicine ,Endocrinology ,Data visualization ,Human Phenotype Ontology ,Databases, Genetic ,Genetics ,medicine ,Humans ,Exome ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Internet ,business.industry ,Information Dissemination ,Computational Biology ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,medicine.disease ,3. Good health ,Data sharing ,Phenotype ,Genome, Mitochondrial ,Female ,business ,030217 neurology & neurosurgery ,Software - Abstract
Success rates for genomic analyses of highly heterogeneous disorders can be greatly improved if a large cohort of patient data is assembled to enhance collective capabilities for accurate sequence variant annotation, analysis, and interpretation. Indeed, molecular diagnostics requires the establishment of robust data resources to enable data sharing that informs accurate understanding of genes, variants, and phenotypes. The "Mitochondrial Disease Sequence Data Resource (MSeqDR) Consortium" is a grass-roots effort facilitated by the United Mitochondria] Disease Foundation to identify and prioritize specific genomic data analysis needs of the global mitochondrial disease clinical and research community. A central Web portal (https://mseqdr.org) facilitates the coherent compilation, organization, annotation, and analysis of sequence data from both nuclear and mitochondrial genomes of individuals and families with suspected mitochondria! disease. This Web portal provides users with a flexible and expandable suite of resources to enable variant-, gene-, and exome-level sequence analysis in a secure, Web-based, and user-friendly fashion. Users can also elect to share data with other MSeqDR Consortium members, or even the general public, either by custom annotation tracks or through the use of a convenient distributed annotation system (DAS) mechanism. A range of data visualization and analysis tools are provided to facilitate user interrogation and understanding of genomic, and ultimately phenotypic, data of relevance to mitochondrial biology and disease. Currently available tools for nuclear and mitochondrial gene analyses include an MSeqDR GBrowse instance that hosts optimized mitochondrial disease and mitochondrial DNA (mtDNA) specific annotation tracks, as well as an MSeqDR locus-specific database (LSDB) that curates variant data on more than 1300 genes that have been implicated in mitochondrial disease and/or encode mitochondria-localized proteins. MSeqDR is integrated with a diverse array of mtDNA data analysis tools that are both freestanding and incorporated into an online exome-level dataset curation and analysis resource (GEM.app) that is being optimized to support needs of the MSeqDR community. In addition, MSeqDR supports mitochondrial disease phenotyping and ontology tools, and provides variant pathogenicity assessment features that enable community review, feedback, and integration with the public ClinVar variant annotation resource. A centralized Web-based informed consent process is being developed, with implementation of a Global Unique Identifier (GUID) system to integrate data deposited on a given individual from different sources. Community-based data deposition into MSeqDR has already begun. Future efforts will enhance capabilities to incorporate phenotypic data that enhance genomic data analyses. MSeqDR will fill the existing void in bioinformatics tools and centralized knowledge that are necessary to enable efficient nuclear and mtDNA genomic data interpretation by a range of shareholders across both clinical diagnostic and research settings. Ultimately, MSeqDR is focused on empowering the global mitochondrial disease community to better define and explore mitochondrial diseases. (C) 2014 Elsevier Inc. All rights reserved.
- Published
- 2015
37. Sequencing of Aspergillus nidulans and comparative analysis with A. fumigatus and A. oryzae
- Author
-
Toshitaka Kumagai, Kay Vienken, Li-Jun Ma, Toshihiro Tanaka, Christina C. Spevak, Paul S. Dyer, Mark L. Farman, Jerzy Jurka, David B. Archer, Claudio Scazzocchio, Christiane Bouchier, Miguel A. Peñalva, Gerhard H. Braus, John H. Doonan, Arnab Pain, Kiyoshi Asai, Eric U. Selker, Reinhard Fischer, James E. Galagan, Seth Purcell, William C. Nierman, Oliver W. Draht, Michelle Momany, Jennifer R. Wortman, Deborah Bell-Pedersen, Bruce W. Birren, Berl R. Oakley, Christina A. Cuomo, David W. Denning, Michael J. Hynes, Jonathan Butler, Su-In Lee, John Clutterbuck, Jae-Hyuk Yu, Christophe d'Enfert, Vladimir V. Kapitonov, Steve Harris, Gustavo H. Goldman, Masayuki Machida, Serafim Batzoglou, Sarah E. Calvo, Mathieu Paoletti, Bruce L. Miller, Matthew S. Sachs, Mark X. Caddick, Silke Busch, Meray Baştürkmen, Michael Freitag, Sam Griffiths-Jones, Stephen A. Osmani, Massachusetts Institute of Technology (MIT), The Institute for Genomic Research (TIGR), Stanford University, Oregon Health & Science University, University of Glasgow, Genetic Information Research Institute, Institut de génétique et microbiologie [Orsay] (IGM), Université Paris-Sud - Paris 11 (UP11)-Centre National de la Recherche Scientifique (CNRS), Department of Plant Pathology, University of Kentucky, University of Nebraska System, Georg-August-University [Göttingen], Biologie et Pathogénicité fongiques, Institut Pasteur [Paris]-Institut National de la Recherche Agronomique (INRA), Institut Pasteur [Paris], Universidade de São Paulo (USP), Department of Biology, University of Minho, The Wellcome Trust Sanger Institute [Cambridge], John Innes Centre, University of Wisconsin-Madison, Max Planck Institute for Terrestrial Microbiology, Max-Planck-Gesellschaft, Institute of Molecular Biology, University of Oregon [Eugene], University of Nottingham, UK (UON), Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Científicas [Madrid] (CSIC), Department of Molecular Genetics, Ohio State University [Columbus] (OSU), Department of Plant Biology, Royal Veterinary and Agricultural University = Kongelige Veterinær- og Landbohøjskole (KVL ), National Institute of Technology [Patna] (NIT), Advanced Industrial Science and technology (AIST), National Institute of Advanced Industrial Science and Technology (AIST), University of Manchester [Manchester], University of Liverpool, Department of Genetics, University of Melbourne, University of Idaho [Moscow, USA], Ohio State University, Partenaires INRAE, University of Kentucky (UK), Georg-August-University = Georg-August-Universität Göttingen, Biologie et Pathogénicité fongiques (BPF), Institut National de la Recherche Agronomique (INRA)-Institut Pasteur [Paris] (IP), Institut Pasteur [Paris] (IP), Universidade de São Paulo = University of São Paulo (USP), University of Minho [Braga], John Innes Centre [Norwich], and Biotechnology and Biological Sciences Research Council (BBSRC)
- Subjects
Proteome ,Molecular Sequence Data ,Genomics ,SEQUENCE DATA ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Regulatory Sequences, Nucleic Acid ,Synteny ,Genome ,Aspergillus fumigatus ,Evolution, Molecular ,Open Reading Frames ,03 medical and health sciences ,GENE REGULATION ,Aspergillus oryzae ,Aspergillus nidulans ,Consensus Sequence ,Humans ,ASPERGILLUS FUMIGATUS ,Gene ,Conserved Sequence ,Phylogeny ,030304 developmental biology ,Genetics ,Whole genome sequencing ,SEQUENCE CONSERVATION ,GENOME EVOLUTION ,0303 health sciences ,Multidisciplinary ,Base Sequence ,biology ,GENOMIC ,030306 microbiology ,Fungal genetics ,Sequence Analysis, DNA ,15. Life on land ,Genes, Mating Type, Fungal ,biology.organism_classification ,ASPERGILLUS ORYZAE ,ASPERGILLUS NIDULANS ,Genome, Fungal - Abstract
International audience; The aspergilli comprise a diverse group of filamentous fungi spanning over 200 million years of evolution. Here we report the genome sequence of the model organism Aspergillus nidulans, and a comparative study with Aspergillus fumigatus, a serious human pathogen, and Aspergillus oryzae, used in the production of sake, miso and soy sauce. Our analysis of genome structure provided a quantitative evaluation of forces driving long-term eukaryotic genome evolution. It also led to an experimentally validated model of mating-type locus evolution, suggesting the potential for sexual reproduction in A. fumigatus and A. oryzae. Our analysis of sequence conservation revealed over 5,000 non-coding regions actively conserved across all three species. Within these regions, we identified potential functional elements including a previously uncharacterized TPP riboswitch and motifs suggesting regulation in filamentous fungi by Puf family genes. We further obtained comparative and experimental evidence indicating widespread translational regulation by upstream open reading frames. These results enhance our understanding of these widely studied fungi as well as provide new insight into eukaryotic genome evolution and gene regulation.
- Published
- 2005
38. The Complete Genome and Proteome of Mycoplasma mobile
- Author
-
Howard C. Berg, Nicole Stange-Thomann, Chinnappa D. Kodira, Robert Nicol, Jonathan Butler, John E. Major, George M. Church, Jacob D. Jaffe, Cherylyn Smith, Jane E. Wilkinson, Nabil Hafez, Sheila Fisher, Shunguang Wang, Sarah E. Calvo, Tim Elkins, Bruce W. Birren, Chad Nusbaum, David DeCaprio, and Michael Fitzgerald
- Subjects
Genetics ,Genome evolution ,Proteome ,Gliding motility ,Molecular Sequence Data ,Computational Biology ,Articles ,Mycoplasma ,Genome project ,Biology ,Physical Chromosome Mapping ,medicine.disease_cause ,Genome ,Phylogenetics ,Horizontal gene transfer ,medicine ,Amino Acid Sequence ,Genome, Bacterial ,Phylogeny ,Genetics (clinical) - Abstract
Although often considered “minimal” organisms, mycoplasmas show a wide range of diversity with respect to host environment, phenotypic traits, and pathogenicity. Here we report the complete genomic sequence and proteogenomic map for the piscine mycoplasma Mycoplasma mobile, noted for its robust gliding motility. For the first time, proteomic data are used in the primary annotation of a new genome, providing validation of expression for many of the predicted proteins. Several novel features were discovered including a long repeating unit of DNA of ∼2435 bp present in five complete copies that are shown to code for nearly identical yet uniquely expressed proteins. M. mobile has among the lowest DNA GC contents (24.9%) and most reduced set of tRNAs of any organism yet reported (28). Numerous instances of tandem duplication as well as lateral gene transfer are evident in the genome. The multiple available complete genome sequences for other motile and immotile mycoplasmas enabled us to use comparative genomic and phylogenetic methods to suggest several candidate genes that might be involved in motility. The results of these analyses leave open the possibility that gliding motility might have arisen independently more than once in the mycoplasma lineage.
- Published
- 2004
39. The genome sequence of the filamentous fungus Neurospora crassa
- Author
-
Matthew G. Endrizzi, Li-Jun Ma, Ian T. Paulsen, Gregory O. Kothe, David B. Jaffe, David E. A. Catcheside, Edward M. Marcotte, Jonathan Butler, Giuseppe Macino, Matthew S. Sachs, David D. Perkins, Jerome Naylor, Oded Yarden, Nick O. Read, Shunguang Wang, Sarah E. Calvo, Reinhard Engels, J. Andrew Berglund, Nicole Stange-Thomann, Robert L. Metzenberg, Stephan Seiler, Scott Kroken, Seth Purcell, Dmitrij Frishman, Ulrich Schulte, Bruce W. Birren, Dayong Qui, Manolis Kamvysselis, Edward L. Braun, Gregory Jedd, Rodolfo Aramayo, Mary Anne Nelson, Sante Gnerre, Carlo Cogoni, Deborah Bell-Pedersen, Rodger B. Voelker, Chad Nusbaum, David Greenberg, Robert J. Pratt, Gertrud Mannhaupt, Bushra Rehman, Carolyn G. Rasmussen, Colin P.C. DeSouza, Michael Plamann, Weixi Li, Evan Mauceli, Daniel J. Ebbole, Katherine A. Borkovich, William Fitzhugh, Svetlana Krystofova, Peter Ianakiev, Claude P. Selitrennikoff, Stephen A. Osmani, Marc J. Orbach, Alice Roy, Jay C. Dunlap, Donald O. Natvig, Robert Barrett, Stephen Rudd, Louise Glass, James E. Galagan, Cord Bielke, Karen Foley, Alan Radford, John A. Kinsey, Michael Freitag, Chuck Staben, Lisa A. Alex, Alex Zelter, Cydney B. Nielsen, Margaret Werner-Washburne, Serge Smirnov, Timothy Elkins, Michael Kamal, Werner Mewes, Eric S. Lander, and Eric U. Selker
- Subjects
Genome evolution ,Genes, Fungal ,Receptors, Cell Surface ,Biology ,Genome ,Neurospora crassa ,Evolution, Molecular ,Multienzyme Complexes ,Gene Duplication ,Gene density ,Calcium Signaling ,Genome size ,Plant Diseases ,Repetitive Sequences, Nucleic Acid ,Genetics ,Multidisciplinary ,fungi ,Sequence Analysis, DNA ,Genome project ,DNA Methylation ,biology.organism_classification ,Heterotrimeric GTP-Binding Proteins ,Mutagenesis ,RNA, Ribosomal ,Multigene Family ,Schizosaccharomyces pombe ,RNA Interference ,Minimal genome ,Diterpenes ,Genome, Fungal ,Signal Transduction - Abstract
Neurospora crassa is a central organism in the history of twentieth-century genetics, biochemistry and molecular biology. Here, we report a high-quality draft sequence of the N. crassa genome. The approximately 40-megabase genome encodes about 10,000 protein-coding genes—more than twice as many as in the fission yeast Schizosaccharomyces pombe and only about 25% fewer than in the fruitfly Drosophila melanogaster. Analysis of the gene set yields insights into unexpected aspects of Neurospora biology including the identification of genes potentially associated with red light photobiology, genes implicated in secondary metabolism, and important differences in Ca21 signalling as compared with plants and animals. Neurospora possesses the widest array of genome defence mechanisms known for any eukaryotic organism, including a process unique to fungi called repeat-induced point mutation (RIP). Genome analysis suggests that RIP has had a profound impact on genome evolution, greatly slowing the creation of new genes through genomic duplication and resulting in a genome with an unusually low proportion of closely related genes.
- Published
- 2003
40. CLYBL is a polymorphic human enzyme with malate synthase and β-methylmalate synthase activity
- Author
-
Vamsi K. Mootha, Sarah E. Calvo, Laura Strittmatter, Zenon Grabarek, Yang Li, and Nathan Nakatsuka
- Subjects
Operon ,Glyoxylate cycle ,Malates ,Mitochondrion ,Malate dehydrogenase ,Substrate Specificity ,Acetyl Coenzyme A ,Malate synthase ,Genetics ,Humans ,Enzyme kinetics ,Molecular Biology ,Genetics (clinical) ,chemistry.chemical_classification ,biology ,ATP synthase ,Malate Synthase ,Glyoxylates ,Oxo-Acid-Lyases ,General Medicine ,Articles ,Enzymes ,Enzyme ,Biochemistry ,chemistry ,biology.protein ,Acyl Coenzyme A - Abstract
CLYBL is a human mitochondrial enzyme of unknown function that is found in multiple eukaryotic taxa and conserved to bacteria. The protein is expressed in the mitochondria of all mammalian organs, with highest expression in brown fat and kidney. Approximately 5% of all humans harbor a premature stop polymorphism in CLYBL that has been associated with reduced levels of circulating vitamin B12. Using comparative genomics, we now show that CLYBL is strongly co-expressed with and co-evolved specifically with other components of the mitochondrial B12 pathway. We confirm that the premature stop polymorphism in CLYBL leads to a loss of protein expression. To elucidate the molecular function of CLYBL, we used comparative operon analysis, structural modeling and enzyme kinetics. We report that CLYBL encodes a malate/β-methylmalate synthase, converting glyoxylate and acetyl-CoA to malate, or glyoxylate and propionyl-CoA to β-methylmalate. Malate synthases are best known for their established role in the glyoxylate shunt of plants and lower organisms and are traditionally described as not occurring in humans. The broader role of a malate/β-methylmalate synthase in human physiology and its mechanistic link to vitamin B12 metabolism remain unknown.
- Published
- 2013
41. The fusarium graminearum genome reveals a link between localized polymorphism and pathogen specialization
- Author
-
David R. Nelson, Hans-Werner Mewes, H. Corby Kistler, Weihong Qi, Kerry O'Donnell, John C. Kennell, Scott E. Baker, Liane R. Gale, Igor V. Tetko, Jon K. Magnuson, Gary J. Muehlbauer, Martin Münsterkötter, Martijn Rep, Karen Hilburn, Jiqiang Yao, B. Gillian Turgeon, Thérèse Ouellet, Hadi Quesneville, Li-Jun Ma, Todd J. Ward, Linda J. Harris, Gerhard Adam, Kim E. Hammond-Kosack, Sarah E. Calvo, Scott Kroken, M. Isabel G. Roncero, Kye Yong Seong, Jin-Rong Xu, Gertrud Mannhaupt, Yueh-Long Chang, Antonio Di Pietro, Ulrich Güldener, Christina A. Cuomo, Evan Mauceli, Rudolf Mitterbauer, John F. Antoniw, Rubella S. Goswami, David DeCaprio, Martin Urban, Cees Waalwijk, Jonathan D. Walton, Sante Gnerre, Frances Trail, Thomas K. Baldwin, Bruce W. Birren, Broad Institute of MIT and Harvard (BROAD INSTITUTE), Harvard Medical School [Boston] (HMS)-Massachusetts Institute of Technology (MIT)-Massachusetts General Hospital [Boston], Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM), Institute for Bioinformatics (MIPS), GSF National Research Center for Environment and Health, Purdue University [West Lafayette], Michigan State University [East Lansing], Michigan State University System, Cornell University [New York], Universidad de Córdoba [Cordoba], Pacific Northwest National Laboratory (PNNL), University of Amsterdam [Amsterdam] (UvA), Universität für Bodenkultur Wien [Vienne, Autriche] (BOKU), Rothamsted Research, University of Minnesota [Twin Cities] (UMN), University of Minnesota System, Agriculture and Agri-Food [Ottawa] (AAFC), U.S. Department of Agriculture - Agricultural Research Service - Cereal Disease Laboratory (USDA), USDA-ARS : Agricultural Research Service, Saint Louis University (SLU), University of Arizona, University of Tennessee Memphis, The University of Tennessee Health Science Center [Memphis] (UTHSC), USDA ARS, National Center for Agricultural Utilization Research (USDA ARS,), Institut Jacques Monod (IJM (UMR_7592)), Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Institute of Bioorganic Chemistry and Photochemistry, National Ukrainian Academy of Sciences, Institute Bioorganic Chemistry an Photochemistry, Plant Research International (PRI), Wageningen University and Research [Wageningen] (WUR), Molecular Plant Pathology (SILS, FNWI), Technische Universität München [München] (TUM), Cornell University, University of Minnesota [Twin Cities], and Wageningen University and Research Centre [Wageningen] (WUR)
- Subjects
localized polymorphism ,MESH: Sequence Analysis, DNA ,neurospora ,Sequence analysis ,Molecular Sequence Data ,Single-nucleotide polymorphism ,Genomics ,Gene mutation ,Biology ,dna ,medicine.disease_cause ,Genome ,Polymorphism, Single Nucleotide ,Evolution, Molecular ,03 medical and health sciences ,MESH: Plant Diseases ,Fusarium ,MESH: Polymorphism, Genetic ,medicine ,Point Mutation ,DNA, Fungal ,Gene ,MESH: Evolution, Molecular ,030304 developmental biology ,Plant Diseases ,MESH: Point Mutation ,Genetics ,0303 health sciences ,Mutation ,MESH: Fusarium ,Multidisciplinary ,Polymorphism, Genetic ,MESH: Molecular Sequence Data ,Biointeracties and Plant Health ,030306 microbiology ,Point mutation ,MESH: Polymorphism, Single Nucleotide ,food and beverages ,Hordeum ,Sequence Analysis, DNA ,[SDV.BIBS]Life Sciences [q-bio]/Quantitative Methods [q-bio.QM] ,MESH: DNA, Fungal ,MESH: Hordeum ,MESH: Genome, Fungal ,Fusarium graminearum genome ,PRI Biointeractions en Plantgezondheid ,Genome, Fungal ,[INFO.INFO-BI]Computer Science [cs]/Bioinformatics [q-bio.QM] - Abstract
We sequenced and annotated the genome of the filamentous fungus Fusarium graminearum , a major pathogen of cultivated cereals. Very few repetitive sequences were detected, and the process of repeat-induced point mutation, in which duplicated sequences are subject to extensive mutation, may partially account for the reduced repeat content and apparent low number of paralogous (ancestrally duplicated) genes. A second strain of F. graminearum contained more than 10,000 single-nucleotide polymorphisms, which were frequently located near telomeres and within other discrete chromosomal segments. Many highly polymorphic regions contained sets of genes implicated in plant-fungus interactions and were unusually divergent, with higher rates of recombination. These regions of genome innovation may result from selection due to interactions of F. graminearum with its plant hosts.
- Published
- 2007
42. Next generation sequencing with copy number variant detection expands the phenotypic spectrum of HSD17B4-deficiency
- Author
-
Nancy G. Slate, Daniel S. Lieber, Vamsi K. Mootha, Sarah E. Calvo, Steven G. Hershman, Katherine B. Sims, and Jeremy D. Schmahmann
- Subjects
Male ,Exome sequencing ,Mitochondrial Diseases ,Phytanic acid ,Gene Dosage ,Case Report ,Compound heterozygosity ,DBP ,chemistry.chemical_compound ,0302 clinical medicine ,Human genetics ,Genetics(clinical) ,Peroxisomal Multifunctional Protein-2 ,Exome ,Genetics (clinical) ,Azoospermia ,Sequence Deletion ,Genetics ,0303 health sciences ,High-Throughput Nucleotide Sequencing ,3. Good health ,Mitochondria ,Phenotype ,Molecular Diagnostic Techniques ,medicine.symptom ,Adult ,Heterozygote ,Ataxia ,HSD17B4 ,DNA Copy Number Variations ,Mitochondrial disease ,Hearing Loss, Sensorineural ,Molecular Sequence Data ,CNV ,Biology ,03 medical and health sciences ,medicine ,Humans ,Multi-system disorders ,Abnormalities, Multiple ,D-bifunctional protein deficiency ,030304 developmental biology ,Peroxisomal defects ,Mitochondrial disorders ,Mendelian disorders ,Perrault syndrome ,Copy number variants ,Cerebellar ataxia ,Base Sequence ,Sequence Analysis, DNA ,medicine.disease ,Molecular biology ,chemistry ,Next-generation sequencing ,030217 neurology & neurosurgery - Abstract
Background: D-bifunctional protein deficiency, caused by recessive mutations in HSD17B4, is a severe, infantile-onset disorder of peroxisomal fatty acid oxidation. Few affected patients survive past two years of age. Compound heterozygous mutations in HSD17B4 have also been reported in two sisters diagnosed with Perrault syndrome (MIM # 233400), who presented in adolescence with ovarian dysgenesis, hearing loss, and ataxia. Case presentation: An adult male presented with cerebellar ataxia, peripheral neuropathy, hearing loss, and azoospermia. The clinical presentation, in combination with biochemical findings in serum, urine, and muscle biopsy, suggested a mitochondrial disorder. Commercial genetic testing of 18 ataxia and mitochondrial disease genes was negative. Targeted exome sequencing followed by analysis of single nucleotide variants and small insertions/deletions failed to reveal a genetic basis of disease. Application of a computational algorithm to infer copy number variants (CNVs) from exome data revealed a heterozygous 12 kb deletion of exons 10–13 of HSD17B4 that was compounded with a rare missense variant (p.A196V) at a highly conserved residue. Retrospective review of patient records revealed mildly elevated ratios of pristanic:phytanic acid and arachidonic:docosahexaenoic acid, consistent with dysfunctional peroxisomal fatty acid oxidation. Conclusion: Our case expands the phenotypic spectrum of HSD17B4-deficiency, representing the first male case reported with infertility. Furthermore, it points to crosstalk between mitochondria and peroxisomes in HSD17B4-deficiency and Perrault syndrome.
- Published
- 2013
43. Targeted exome sequencing of suspected mitochondrial disorders
- Author
-
Gerard T. Berry, Kristy Shanahan, Nancy G. Slate, David R. Thorburn, Mark L. Borowsky, Daniel S. Lieber, Shangtao Liu, Vamsi K. Mootha, Nina B. Gold, Steven G. Hershman, Katherine B. Sims, Sarah E. Calvo, Brad Chapman, David M. Mueller, and Jeremy D. Schmahmann
- Subjects
Adult ,Male ,Mitochondrial DNA ,Mitochondrial Diseases ,Adolescent ,Mitochondrial disease ,Molecular Sequence Data ,NDUFV1 ,Biology ,DNA, Mitochondrial ,Article ,Young Adult ,medicine ,Humans ,Exome ,Genetic Predisposition to Disease ,Amino Acid Sequence ,Child ,Gene ,Exome sequencing ,Genetics ,Genetic heterogeneity ,Infant, Newborn ,Infant ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,Pedigree ,Child, Preschool ,Gene Targeting ,DPYD ,Female ,Neurology (clinical) - Abstract
Objective: To evaluate the utility of targeted exome sequencing for the molecular diagnosis of mitochondrial disorders, which exhibit marked phenotypic and genetic heterogeneity. Methods: We considered a diverse set of 102 patients with suspected mitochondrial disorders based on clinical, biochemical, and/or molecular findings, and whose disease ranged from mild to severe, with varying age at onset. We sequenced the mitochondrial genome (mtDNA) and the exons of 1,598 nuclear-encoded genes implicated in mitochondrial biology, mitochondrial disease, or monogenic disorders with phenotypic overlap. We prioritized variants likely to underlie disease and established molecular diagnoses in accordance with current clinical genetic guidelines. Results: Targeted exome sequencing yielded molecular diagnoses in established disease loci in 22% of cases, including 17 of 18 (94%) with prior molecular diagnoses and 5 of 84 (6%) without. The 5 new diagnoses implicated 2 genes associated with canonical mitochondrial disorders ( NDUFV1 , POLG2 ), and 3 genes known to underlie other neurologic disorders ( DPYD , KARS , WFS1 ), underscoring the phenotypic and biochemical overlap with other inborn errors. We prioritized variants in an additional 26 patients, including recessive, X-linked, and mtDNA variants that were enriched 2-fold over background and await further support of pathogenicity. In one case, we modeled patient mutations in yeast to provide evidence that recessive mutations in ATP5A1 can underlie combined respiratory chain deficiency. Conclusion: The results demonstrate that targeted exome sequencing is an effective alternative to the sequential testing of mtDNA and individual nuclear genes as part of the investigation of mitochondrial disease. Our study underscores the ongoing challenge of variant interpretation in the clinical setting.
- Published
- 2013
44. Next-generation sequencing reveals DGUOK mutations in adult patients with mitochondrial DNA multiple deletions
- Author
-
Salvatore DiMauro, Michela Ranieri, Nereo Bresolin, Vamsi K. Mootha, Dario Ronchi, Stefania Corti, Caterina Garone, Francesca Magri, Mafalda Rizzuti, Purificacion Gutierrez Rios, Luisa Villa, Michela Ripolone, Monica Sciacco, Giacomo P. Comi, Sarah E. Calvo, Maurizio Moggio, Andreina Bordoni, Ronchi D., Garone C., Bordoni A., Gutierrez Rios P., Calvo S.E., Ripolone M., Ranieri M., Rizzuti M., Villa L., Magri F., Corti S., Bresolin N., Mootha V.K., Moggio M., Dimauro S., Comi G.P., and Sciacco M.
- Subjects
Adult ,Male ,Mitochondrial DNA ,Mitochondrial Diseases ,Mitochondrial disease ,Molecular Sequence Data ,autosomal recessive progressive external ophthalmoplegia ,Mitochondrion ,Biology ,DGUOK ,medicine.disease_cause ,DNA, Mitochondrial ,Polymorphism, Single Nucleotide ,multiple mitochondrial DNA deletion ,DNA sequencing ,Mitochondrial myopathy ,mitochondrial DNA instability ,Mitochondrial Disease ,medicine ,Humans ,Muscle, Skeletal ,Aged ,Genetics ,Aged, 80 and over ,Mutation ,Base Sequence ,Multiple mitochondrial DNA deletions ,Original Articles ,Middle Aged ,medicine.disease ,Phosphotransferases (Alcohol Group Acceptor) ,Female ,Neurology (clinical) ,Gene Deletion ,Human - Abstract
The molecular diagnosis of mitochondrial disorders still remains elusive in a large proportion of patients, but advances in next generation sequencing are significantly improving our chances to detect mutations even in sporadic patients. Syndromes associated with mitochondrial DNA multiple deletions are caused by different molecular defects resulting in a wide spectrum of predominantly adult-onset clinical presentations, ranging from progressive external ophthalmoplegia to multi-systemic disorders of variable severity. The mutations underlying these conditions remain undisclosed in half of the affected subjects. We applied next-generation sequencing of known mitochondrial targets (MitoExome) to probands presenting with adult-onset mitochondrial myopathy and harbouring mitochondrial DNA multiple deletions in skeletal muscle. We identified autosomal recessive mutations in the DGUOK gene (encoding mitochondrial deoxyguanosine kinase), which has previously been associated with an infantile hepatocerebral form of mitochondrial DNA depletion. Mutations in DGUOK occurred in five independent subjects, representing 5.6% of our cohort of patients with mitochondrial DNA multiple deletions, and impaired both muscle DGUOK activity and protein stability. Clinical presentations were variable, including mitochondrial myopathy with or without progressive external ophthalmoplegia, recurrent rhabdomyolysis in a young female who had received a liver transplant at 9 months of age and adult-onset lower motor neuron syndrome with mild cognitive impairment. These findings reinforce the concept that mutations in genes involved in deoxyribonucleotide metabolism can cause diverse clinical phenotypes and suggest that DGUOK should be screened in patients harbouring mitochondrial DNA deletions in skeletal muscle. © 2012 The Author (2012). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: journals.permissions@oup.com.
- Published
- 2012
45. Loss-of-function mutations in MGME1 impair mtDNA replication and cause multisystemic mitochondrial disease
- Author
-
Arcangela Iuso, Thomas Meitinger, Salvatore DiMauro, Holger Prokisch, Vamsi K. Mootha, Viktoriya Peeva, Giacomo P. Comi, Maurizio Moggio, Katharina Danhauser, Dario Ronchi, Michal Minczuk, Sarah E. Calvo, Joanna Rorbach, Thomas Klopstock, Gábor Zsurka, Wolfram S. Kunz, Kerstin Hallmann, Susanne Schöler, Tim M. Strom, Tobias B. Haack, Monica Sciacco, Catarina M. Quinzii, Thomas Wieland, Cornelia Kornblum, and Thomas J. Nicholls
- Subjects
Exonuclease ,DNA Replication ,Models, Molecular ,Mitochondrial DNA ,Mitochondrial Diseases ,Mitochondrial disease ,Molecular Sequence Data ,Biology ,Human mitochondrial genetics ,DNA, Mitochondrial ,Article ,chemistry.chemical_compound ,Exonuclease 1 ,Genetics ,medicine ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,DNA Primers ,Base Sequence ,DNA replication ,Mitochondrial genome maintenance ,Sequence Analysis, DNA ,medicine.disease ,Molecular biology ,Exodeoxyribonucleases ,Gene Components ,chemistry ,Codon, Nonsense ,biology.protein ,DNA ,HeLa Cells - Abstract
Known disease mechanisms in mitochondrial DNA (mtDNA) maintenance disorders alter either the mitochondrial replication machinery (POLG1, POLG22 and C10orf23) or the biosynthesis pathways of deoxyribonucleoside 5′-triphosphates for mtDNA synthesis4–11. However, in many of these disorders, the underlying genetic defect has not yet been discovered. Here, we identified homozygous nonsense and missense mutations in the orphan gene C20orf72 in three families with a mitochondrial syndrome characterized by external ophthalmoplegia, emaciation, and respiratory failure. Muscle biopsies showed mtDNA depletion and multiple mtDNA deletions. C20orf72, hereafter MGME1 (mitochondrial genome maintenance exonuclease 1), encodes a mitochondrial RecB-type exonuclease belonging to the PD-(D/E)XK nuclease superfamily. We demonstrate that MGME1 cleaves single-stranded DNA and processes DNA flap substrates. Upon chemically induced mtDNA depletion, patient fibroblasts fail to repopulate. They also accumulate intermediates of stalled replication and show increased levels of 7S DNA, as do MGME1-depleted cells. Hence, we show that MGME1-mediated mtDNA processing is essential for mitochondrial genome maintenance.
- Published
- 2012
46. Evolutionary diversity of the mitochondrial calcium uniporter
- Author
-
Vamsi K. Mootha, Sarah E. Calvo, and Alexander G. Bick
- Subjects
Proteome ,Bacterial genome size ,Mitochondrion ,Genome ,Mitochondrial Membrane Transport Proteins ,Article ,Evolution, Molecular ,Mitochondrial membrane transport protein ,Bacterial Proteins ,Phylogenetics ,Calcium-binding protein ,Animals ,Humans ,Uniporter ,Cation Transport Proteins ,Phylogeny ,Multidisciplinary ,biology ,Voltage-dependent calcium channel ,Bacteria ,Calcium-Binding Proteins ,Eukaryota ,Cell biology ,Mitochondria ,Protein Structure, Tertiary ,biology.protein ,Calcium Channels - Abstract
Calcium uptake into mitochondria occurs via a recently identified ion channel called the uniporter. Here, we characterize the phylogenomic distribution of the uniporter’s membrane-spanning pore subunit (MCU) and regulatory partner (MICU1). Homologs of both components tend to co-occur in all major branches of eukaryotic life, but both have been lost along certain protozoan and fungal lineages. Several bacterial genomes also contain putative MCU homologs that may represent prokaryotic calcium channels. The analyses indicate that the uniporter may have been an early feature of mitochondria.
- Published
- 2012
47. The molecular basis of human complex I deficiency
- Author
-
Sarah E. Calvo, Elena J. Tucker, David R. Thorburn, and Alison G. Compton
- Subjects
medicine.medical_specialty ,Mitochondrial Diseases ,Ataxia ,Nuclear gene ,Mitochondrial disease ,Clinical Biochemistry ,Biology ,DNA, Mitochondrial ,Biochemistry ,Locus heterogeneity ,Molecular genetics ,Genetics ,medicine ,Humans ,Missense mutation ,Allele ,Molecular Biology ,Gene ,Electron Transport Complex I ,Computational Biology ,Cell Biology ,medicine.disease ,Protein Subunits ,Genes ,Mutation ,medicine.symptom ,Metabolism, Inborn Errors - Abstract
Disorders of oxidative phosphorylation (OXPHOS) have a birth prevalence of ∼1/5,000 and are the most common inborn errors of metabolism. The most common OXPHOS disorder is complex I deficiency. Patients with complex I deficiency present with variable symptoms, such as muscle weakness, cardiomyopathy, developmental delay or regression, blindness, seizures, failure to thrive, liver dysfunction or ataxia. Molecular diagnosis of patients with complex I deficiency is a challenging task due to the clinical heterogeneity of patients and the large number of candidate disease genes, both nuclear-encoded and mitochondrial DNA (mtDNA)-encoded. In this review, we have thoroughly surveyed the literature to identify 149 patients described with both isolated complex I deficiency and pathogenic mutations within nuclear genes. In total, 115 different pathogenic mutations have been reported in 22 different nuclear genes encoding complex I subunits or assembly factors, highlighting the allelic and locus heterogeneity of this disorder. Missense mutations predominate in genes encoding core subunits and some assembly factors while null-type mutations are common in the genes encoding supernumerary subunits and other assembly factors. Despite developments in molecular technology, many patients do not receive molecular diagnosis and no gene has yet been identified that accounts for more than 5% of cases, suggesting that there are likely many disease genes that await discovery.© 2011 IUBMB Life, 63(9): 669–677, 2011
- Published
- 2011
48. FOXRED1, encoding an FAD-dependent oxidoreductase complex-I-specific molecular chaperone, is mutated in infantile-onset mitochondrial encephalopathy
- Author
-
Shamima Rahman, Elisa Fassone, Alistair T. Pagnamenta, Waseem Qasim, Vamsi K. Mootha, Maria Bitner-Glindzicz, Jan-Willem Taanman, Andrew J. Duncan, Sarah E. Calvo, Tatjana Holand, Paul Rutland, and Michael I. Sadowski
- Subjects
Oxidoreductase complex ,Male ,Models, Molecular ,Mitochondrial disease ,DNA Mutational Analysis ,Molecular Sequence Data ,Respiratory chain ,Biology ,Mitochondrion ,medicine.disease_cause ,Mitochondrial Encephalomyopathies ,Genetics ,medicine ,Humans ,Amino Acid Sequence ,Gene Silencing ,RNA, Messenger ,Inner mitochondrial membrane ,Child ,Molecular Biology ,Genetics (clinical) ,Mutation ,Electron Transport Complex I ,Base Sequence ,Genetic Complementation Test ,Homozygote ,Lentivirus ,Computational Biology ,Infant ,General Medicine ,Articles ,medicine.disease ,Mitochondria ,Protein Transport ,Gene Expression Regulation ,Child, Preschool ,Flavin-Adenine Dinucleotide ,Corrigendum ,Molecular Chaperones ,Subcellular Fractions - Abstract
Complex I is the first and largest enzyme in the respiratory chain and is located in the inner mitochondrial membrane. Complex I deficiency is the most commonly reported mitochondrial disorder presenting in childhood, but the molecular basis of most cases remains elusive. We describe a patient with complex I deficiency caused by mutation of the molecular chaperone FOXRED1. A combined homozygosity mapping and bioinformatics approach in a consanguineous Iranian-Jewish pedigree led to the identification of a homozygous mutation in FOXRED1 in a child who presented with infantile-onset encephalomyopathy. Silencing of FOXRED1 in human fibroblasts resulted in reduced complex I steady-state levels and activity, while lentiviral-mediated FOXRED1 transgene expression rescued complex I deficiency in the patient fibroblasts. This FAD-dependent oxidoreductase, which has never previously been associated with human disease, is now shown to be a complex I-specific molecular chaperone. The discovery of the c.1054C>T; p.R352W mutation in the FOXRED1 gene is a further contribution towards resolving the complex puzzle of the genetic basis of human mitochondrial disease.
- Published
- 2010
49. High-throughput, pooled sequencing identifies mutations in NUBPL and FOXRED1 in human complex I deficiency
- Author
-
Vamsi K. Mootha, Michelle C Redman, Olga Goldberger, Damien L. Bruno, Sarah E. Calvo, Candace Guiducci, Manuel A. Rivas, Esko Wiltshire, Elena J. Tucker, Noël P. Burtt, David R. Thorburn, David Altshuler, Callum Wilson, Mark J. Daly, Gabriel Crawford, Alison G. Compton, Stacey Gabriel, and Denise M. Kirby
- Subjects
Candidate gene ,Nuclear gene ,Mitochondrial Diseases ,Sequence analysis ,Blotting, Western ,Gene Dosage ,Biology ,medicine.disease_cause ,Deep sequencing ,Article ,Mitochondrial Proteins ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Humans ,RNA, Messenger ,Exome sequencing ,Genetic Association Studies ,030304 developmental biology ,0303 health sciences ,Mutation ,Electron Transport Complex I ,Reverse Transcriptase Polymerase Chain Reaction ,Sequence Analysis, DNA ,3. Good health ,Complementation ,Mitochondrial respiratory chain ,Case-Control Studies ,030217 neurology & neurosurgery - Abstract
Discovering the molecular basis of mitochondrial respiratory chain disease is challenging given the large number of both mitochondrial and nuclear genes that are involved. We report a strategy of focused candidate gene prediction, high-throughput sequencing and experimental validation to uncover the molecular basis of mitochondrial complex I disorders. We created seven pools of DNA from a cohort of 103 cases and 42 healthy controls and then performed deep sequencing of 103 candidate genes to identify 151 rare variants that were predicted to affect protein function. We established genetic diagnoses in 13 of 60 previously unsolved cases using confirmatory experiments, including cDNA complementation to show that mutations in NUBPL and FOXRED1 can cause complex I deficiency. Our study illustrates how large-scale sequencing, coupled with functional prediction and experimental validation, can be used to identify causal mutations in individual cases.
- Published
- 2010
50. Upstream open reading frames cause widespread reduction of protein expression and are polymorphic among humans
- Author
-
Sarah E. Calvo, Vamsi K. Mootha, and David J. Pagliarini
- Subjects
Untranslated region ,Genetics ,Translation reinitiation ,Multidisciplinary ,Polymorphism, Genetic ,Base Sequence ,Molecular Sequence Data ,Biology ,Biological Sciences ,Open reading frame ,Open Reading Frames ,Start codon ,Protein Biosynthesis ,Upstream open reading frame ,Mutation ,Coding region ,Humans ,Disease ,5' Untranslated Regions ,Post-transcriptional regulation ,Gene - Abstract
Upstream ORFs (uORFs) are mRNA elements defined by a start codon in the 5′ UTR that is out-of-frame with the main coding sequence. Although uORFs are present in approximately half of human and mouse transcripts, no study has investigated their global impact on protein expression. Here, we report that uORFs correlate with significantly reduced protein expression of the downstream ORF, based on analysis of 11,649 matched mRNA and protein measurements from 4 published mammalian studies. Using reporter constructs to test 25 selected uORFs, we estimate that uORFs typically reduce protein expression by 30–80%, with a modest impact on mRNA levels. We additionally identify polymorphisms that alter uORF presence in 509 human genes. Finally, we report that 5 uORF-altering mutations, detected within genes previously linked to human diseases, dramatically silence expression of the downstream protein. Together, our results suggest that uORFs influence the protein expression of thousands of mammalian genes and that variation in these elements can influence human phenotype and disease.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.