524 results on '"Clarke, Catherine F"'
Search Results
152. Primary Coenzyme Q Deficiency in Pdss2 Mutant Mice Causes Isolated Renal Disease
- Author
-
Peng, Min, primary, Falk, Marni J., additional, Haase, Volker H., additional, King, Rhonda, additional, Polyak, Erzsebet, additional, Selak, Mary, additional, Yudkoff, Marc, additional, Hancock, Wayne W., additional, Meade, Ray, additional, Saiki, Ryoichi, additional, Lunceford, Adam L., additional, Clarke, Catherine F., additional, and L. Gasser, David, additional
- Published
- 2008
- Full Text
- View/download PDF
153. Coq2p function in the Saccharomyces cerevisiae coenzyme Q biosynthetic pathway
- Author
-
Marbois, Beth Noelle, primary, Xie, Letian, additional, Shi, Yuchen, additional, Hyman, Kyle, additional, and Clarke, Catherine F., additional
- Published
- 2008
- Full Text
- View/download PDF
154. Investigation of the Interacting Partners of Yeast Coq6: A Component of the Multienzyme Complex Required for Coenzyme Q Biosynthesis
- Author
-
Osonkie, Odi, primary and Clarke, Catherine F, additional
- Published
- 2006
- Full Text
- View/download PDF
155. Ubiquinone is not required for proton conductance by uncoupling protein 1 in yeast mitochondria
- Author
-
ESTEVES, Telma C., primary, ECHTAY, Karim S., additional, JONASSEN, Tanya, additional, CLARKE, Catherine F., additional, and BRAND, Martin D., additional
- Published
- 2004
- Full Text
- View/download PDF
156. A tRNATRP gene mediates the suppression of cbs2-223 previously attributed to ABC1/COQ8
- Author
-
Hsieh, Edward J., primary, Dinoso, Jason B., additional, and Clarke, Catherine F., additional
- Published
- 2004
- Full Text
- View/download PDF
157. Coenzyme Q6 and iron reduction are responsible for the extracellular ascorbate stabilization at the plasma membrane of Saccharomyces cerevisiae
- Author
-
Dirección General de Enseñanza Superior e Investigación Científica (España), Ministerio de Educación y Ciencia (España), National Institutes of Health (US), Santos-Ocaña, Carlos, Córdoba, Francisco, Crane, Frederick L., Clarke, Catherine F., Navas, Plácido, Dirección General de Enseñanza Superior e Investigación Científica (España), Ministerio de Educación y Ciencia (España), National Institutes of Health (US), Santos-Ocaña, Carlos, Córdoba, Francisco, Crane, Frederick L., Clarke, Catherine F., and Navas, Plácido
- Abstract
Yeast plasma membrane contains an electron transport system that maintains ascorbate in its reduced form in the apoplast. Reduction of ascorbate free radical by this system is comprised of two activities, one of them dependent on coenzyme Q6 (CoQ6). Strains with defects in CoQ6 synthesis exhibit decreased capacity for ascorbate stabilization compared with wild type or with atp2 or cor1 respiratory-deficient mutant strains. Both CoQ6 content in plasma membranes and ascorbate stabilization were increased during log phase growth. The addition of exogenous CoQ6 to whole cells resulted in its incorporation in the plasma membrane, produced levels of CoQ6 in the coq3 mutant strain that were 2-fold higher than in the wild type, and increased ascorbate stabilization activity in both strains, although it was higher in the coq3 mutant than in wild type. Other antioxidants, such as benzoquinone or alpha-tocopherol, did not change ascorbate stabilization. The CoQ6-independent reduction of ascorbate free radical was not due to copper uptake, pH changes or to the presence of CoQ6 biosynthetic intermediates, but decreased to undetectable levels when coq3 mutant strains were cultured in media supplemented with ferric iron. Plasma membrane CoQ6 levels were unchanged by either the presence or absence of iron in wild type, atp2, or cor1 strains. Ascorbate stabilization appears to be a function of the yeast plasma membrane, which is partially based on an electron transfer chain in which CoQ6 is the central electron carrier, whereas the remainder is independent of CoQ6 and other antioxidants but is dependent on the iron-regulated ferric reductase complex.
- Published
- 1998
158. Genetic evidence for coenzyme Q requirement in plasma membrane electron transport
- Author
-
National Institutes of Health (US), Universidad de Córdoba (España), Santos-Ocaña, Carlos, Villalba, José M., Córdoba, Francisco, Padilla, Sergio, Crane, Frederick L., Clarke, Catherine F., Navas, Plácido, National Institutes of Health (US), Universidad de Córdoba (España), Santos-Ocaña, Carlos, Villalba, José M., Córdoba, Francisco, Padilla, Sergio, Crane, Frederick L., Clarke, Catherine F., and Navas, Plácido
- Abstract
Plasma membranes isolated from wild-type Saccharomyces cerevisiae crude membrane fractions catalyzed NADH oxidation using a variety of electron acceptors, such as ferricyanide, cytochrome c, and ascorbate free radical. Plasma membranes from the deletion mutant strain coq3delta, defective in coenzyme Q (ubiquinone) biosynthesis, were completely devoid of coenzyme Q6 and contained greatly diminished levels of NADH-ascorbate free radical reductase activity (about 10% of wild-type yeasts). In contrast, the lack of coenzyme Q6 in these membranes resulted in only a partial inhibition of either the ferricyanide or cytochrome-c reductase. Coenzyme Q dependence of ferricyanide and cytochrome-c reductases was based mainly on superoxide generation by one-electron reduction of quinones to semiquinones. Ascorbate free radical reductase was unique because it was highly dependent on coenzyme Q and did not involve superoxide since it was not affected by superoxide dismutase (SOD). Both coenzyme Q6 and NADH-ascorbate free radical reductase were rescued in plasma membranes derived from a strain obtained by transformation of the coq3delta strain with a single-copy plasmid bearing the wild type COQ3 gene and in plasma membranes isolated form the coq3delta strain grown in the presence of coenzyme Q6. The enzyme activity was inhibited by the quinone antagonists chloroquine and dicumarol, and after membrane solubilization with the nondenaturing detergent Zwittergent 3-14. The various inhibitors used did not affect residual ascorbate free radical reductase of the coq3delta strain. Ascorbate free radical reductase was not altered significantly in mutants atp2delta and cor1delta which are also respiration-deficient but not defective in ubiquinone biosynthesis, demonstrating that the lack of ascorbate free radical reductase in coq3delta mutants is related solely to the inability to synthesize ubiquinone and not to the respiratory-defective phenotype. For the first time, our results provide ge
- Published
- 1998
159. P5 - TARGETING FREE RADICALS: A VALID THERAPEUTIC APPROACH FOR HUNTINGTON’S DISEASE?
- Author
-
Chesselet, Marie-Françoise, Relano-Gines, Aroa, Clarke, Catherine F., and Shchepinov, Mikhail S
- Published
- 2014
- Full Text
- View/download PDF
160. Extension of Life-Span in Caenorhabditis elegans by a Diet Lacking Coenzyme Q
- Author
-
Larsen, Pamela L., primary and Clarke, Catherine F., additional
- Published
- 2002
- Full Text
- View/download PDF
161. A dietary source of coenzyme Q is essential for growth of long-lived Caenorhabditis elegans clk-1 mutants
- Author
-
Jonassen, Tanya, primary, Larsen, Pamela L., additional, and Clarke, Catherine F., additional
- Published
- 2001
- Full Text
- View/download PDF
162. Identification of Escherichia coli ubiB , a Gene Required for the First Monooxygenase Step in Ubiquinone Biosynthesis
- Author
-
Poon, Wayne W., primary, Davis, Diana E., additional, Ha, Huan T., additional, Jonassen, Tanya, additional, Rather, Philip N., additional, and Clarke, Catherine F., additional
- Published
- 2000
- Full Text
- View/download PDF
163. Characterization ofSaccharomyces cerevisiaeubiquinone-deficient mutants
- Author
-
Schultz, Jeffery R., primary and Clarke, Catherine F., additional
- Published
- 1999
- Full Text
- View/download PDF
164. Coenzyme Q6 and Iron Reduction Are Responsible for the Extracellular Ascorbate Stabilization at the Plasma Membrane of Saccharomyces cerevisiae
- Author
-
Santos-Ocaña, Carlos, primary, Córdoba, Francisco, additional, Crane, Frederick L., additional, Clarke, Catherine F., additional, and Navas, Plácido, additional
- Published
- 1998
- Full Text
- View/download PDF
165. Yeast Clk-1 Homologue (Coq7/Cat5) Is a Mitochondrial Protein in Coenzyme Q Synthesis
- Author
-
Jonassen, Tanya, primary, Proft, Markus, additional, Randez-Gil, Francisca, additional, Schultz, Jeffery R., additional, Marbois, B. Noelle, additional, Entian, Karl-Dieter, additional, and Clarke, Catherine F., additional
- Published
- 1998
- Full Text
- View/download PDF
166. Characterization of the COQ5 Gene from Saccharomyces cerevisiae EVIDENCE FOR A C-METHYLTRANSFERASE IN UBIQUINONE BIOSYNTHESIS
- Author
-
Barkovich, Robert J., primary, Shtanko, Andrey, additional, Shepherd, Jennifer A., additional, Lee, Peter T., additional, Myles, David C., additional, Tzagoloff, Alexander, additional, and Clarke, Catherine F., additional
- Published
- 1997
- Full Text
- View/download PDF
167. Sensitivity to treatment with polyunsaturated fatty acids is a general characteristic of the ubiquinone-deficient yeast coq mutants
- Author
-
Poon, Wayne W., primary, Do, Thai Q., additional, Noelle Marbois, B., additional, and Clarke, Catherine F., additional
- Published
- 1997
- Full Text
- View/download PDF
168. Isolation and Sequencing of the RatCoq7Gene and the Mapping of MouseCoq7to Chromosome 7
- Author
-
Jonassen, Tanya, primary, Marbois, B.Noelle, additional, Kim, Lena, additional, Chin, Arnold, additional, Xia, Yu-Rong, additional, Lusis, Aldons J., additional, and Clarke, Catherine F., additional
- Published
- 1996
- Full Text
- View/download PDF
169. The biosynthesis of ubiquinone: Synthesis and enzymatic modification of biosynthetic precursors
- Author
-
Shepherd, Jennifer A., primary, Poon, Wayne W., additional, Myles, David C., additional, and Clarke, Catherine F., additional
- Published
- 1996
- Full Text
- View/download PDF
170. The COQ7 Gene Encodes a Protein in Saccharomyces cerevisiae Necessary for Ubiquinone Biosynthesis
- Author
-
Marbois, B. Noelle, primary and Clarke, Catherine F., additional
- Published
- 1996
- Full Text
- View/download PDF
171. 3-Hexaprenyl-4-hydroxybenzoic acid forms a predominant intermediate pool in ubiquinone biosynthesis in Saccharomyces cerevisiae
- Author
-
Poon, Wayne W., primary, Marbois, B.Noelle, additional, Faull, Kym F., additional, and Clarke, Catherine F., additional
- Published
- 1995
- Full Text
- View/download PDF
172. Resveratrol and para-coumarate serve as ring precursors for coenzyme Q biosynthesis[S]
- Author
-
Xie, Letian X., Williams, Kevin J., He, Cuiwen H., Weng, Emily, Khong, San, Rose, Tristan E., Kwon, Ohyun, Bensinger, Steven J., Marbois, Beth N., and Clarke, Catherine F.
- Abstract
Coenzyme Q (Q or ubiquinone) is a redox-active polyisoprenylated benzoquinone lipid essential for electron and proton transport in the mitochondrial respiratory chain. The aromatic ring 4-hydroxybenzoic acid (4HB) is commonly depicted as the sole aromatic ring precursor in Q biosynthesis despite the recent finding that para-aminobenzoic acid (pABA) also serves as a ring precursor in Saccharomyces cerevisiaeQ biosynthesis. In this study, we employed aromatic 13C6-ring-labeled compounds including 13C6-4HB, 13C6-pABA, 13C6-resveratrol, and 13C6-coumarate to investigate the role of these small molecules as aromatic ring precursors in Q biosynthesis in Escherichia coli, S. cerevisiae, and human and mouse cells. In contrast to S. cerevisiae, neither E. colinor the mammalian cells tested were able to form 13C6-Q when cultured in the presence of 13C6-pABA. However, E. colicells treated with 13C6-pABA generated 13C6-ring-labeled forms of 3-octaprenyl-4-aminobenzoic acid, 2-octaprenyl-aniline, and 3-octaprenyl-2-aminophenol, suggesting UbiA, UbiD, UbiX, and UbiI are capable of using pABA or pABA-derived intermediates as substrates. E. coli, S. cerevisiae,and human and mouse cells cultured in the presence of 13C6-resveratrol or 13C6-coumarate were able to synthesize 13C6-Q. Future evaluation of the physiological and pharmacological responses to dietary polyphenols should consider their metabolism to Q.
- Published
- 2015
- Full Text
- View/download PDF
173. Cloning of a rat cDNA encoding dihydroxypolyprenylbenzoate methyltransferase by functional complementation of a Saccharomyces cerevisiae mutant deficient in ubiquinone biosynthesis
- Author
-
Noelle Marbois, B., primary, Hsu, Adam, additional, Pillai, Rajeswari, additional, Colicelli, John, additional, and Clarke, Catherine F., additional
- Published
- 1994
- Full Text
- View/download PDF
174. Focal segmental glomerulosclerosis is associated with a PDSS2 haplotype and, independently, with a decreased content of coenzyme Q10.
- Author
-
Gasser, David L., Winkler, Cheryl A., Min Peng, Ping An, McKenzie, Louise M., Kirk, Gregory D., Yuchen Shi, Letian X. Xie, Marbois, Beth N., Clarke, Catherine F., and Kopp, Jeffrey B.
- Subjects
FOCAL segmental glomerulosclerosis ,PYROPHOSPHATES ,TUMOR suppressor proteins ,UBIQUINONES ,NEPHROTIC syndrome ,SINGLE nucleotide polymorphisms - Abstract
Gasser DL, Winkler CA, Peng M, An P, McKenzie LM, Kirk GD, Shi Y, Xie LX, Marbois BN, Clarke CF, Kopp JB. Focal segmental glomerulosclerosis is associated with a PDSS2 haplotype and, independently, with a decreased content of coenzyme Q10. Am J Physiol Renal Physiol 305: F1228-F1238, 2013. First published August 7, 2013; doi:10.1152/ajprenal.00143.2013.--Focal segmental glomerulosclerosis (FSGS) and collapsing glomerulopathy are common causes of nephrotic syndrome. Variants in >20 genes, including genes critical for mitochondrial function, have been associated with these podocyte diseases. One such gene, PDSS2, is required for synthesis of the decaprenyl tail of coenzyme Q
10 (Q10 ) in humans. The mouse gene Pdss2 is mutated in the kd/kd mouse model of collapsing glomerulopathy. We examined the hypothesis that human PDSS2 polymorphisms are associated with podocyte diseases. We genotyped 377 patients with primary FSGS or collapsing glomerulopathy, together with 900 controls, for 9 single-nucleotide polymorphisms in the PDSS2 gene in a case-control study. Subjects included 247 African American (AA) and 130 European American (EA) patients and 641 AA and 259 EA controls. Among EAs, a pair of proxy SNPs was significantly associated with podocyte disease, and patients homozygous for one PDSS2 haplotype had a strongly increased risk for podocyte disease. By contrast, the distribution of PDSS2 genotypes and haplotypes was similar in AA patients and controls. Thus a PDSS2 haplotype, which has a frequency of 13% in the EA control population and a homozygote frequency of 1.2%, is associated with a significantly increased risk for FSGS and collapsing glomerulopathy in EAs. Lymphoblastoid cell lines from FSGS patients had significantly less Q10 than cell lines from controls; contrary to expectation, this finding was independent of PDSS2 haplotype. These results suggest that FSGS patients have Q10 deficiency and that this deficiency is manifested in patient-derived lymphoblastoid cell lines. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
175. 176th ENMC International Workshop: Diagnosis and treatment of coenzyme Q10 deficiency
- Author
-
Rahman, Shamima, Clarke, Catherine F., and Hirano, Michio
- Published
- 2012
- Full Text
- View/download PDF
176. Delayed accumulation of intestinal coliform bacteria enhances life span and stress resistance in Caenorhabditis elegans fed respiratory deficient E. coli.
- Author
-
Gomez, Fernando, Monsalve, Gabriela C., Tse, Vincent, Ryoichi Saiki, Weng, Emily, Lee, Laura, Srinivasan, Chandra, Frand, Alison R., and Clarke, Catherine F.
- Subjects
ENTEROBACTERIACEAE ,CAENORHABDITIS elegans ,LIFE spans ,FLUORESCENCE microscopy ,ESCHERICHIA coli - Abstract
Background: Studies with the nematode model Caenorhabditis elegans have identified conserved biochemical pathways that act to modulate life span. Life span can also be influenced by the composition of the intestinal microbiome, and C. elegans life span can be dramatically influenced by its diet of Escherichia coli. Although C. elegans is typically fed the standard OP50 strain of E. coli, nematodes fed E. coli strains rendered respiratory deficient, either due to a lack coenzyme Q or the absence of ATP synthase, show significant life span extension. Here we explore the mechanisms accounting for the enhanced nematode life span in response to these diets. Results: The intestinal load of E. coli was monitored by determination of worm-associated colony forming units (cfu/worm or coliform counts) as a function of age. The presence of GFP-expressing E. coli in the worm intestine was also monitored by fluorescence microscopy. Worms fed the standard OP50 E. coli strain have high cfu and GFP-labeled bacteria in their guts at the L4 larval stage, and show saturated coliform counts by day five of adulthood. In contrast, nematodes fed diets of respiratory deficient E. coli lacking coenzyme Q lived significantly longer and failed to accumulate bacteria within the lumen at early ages. Animals fed bacteria deficient in complex V showed intermediate coliform numbers and were not quite as long-lived. The results indicate that respiratory deficient Q-less E. coli are effectively degraded in the early adult worm, either at the pharynx or within the intestine, and do not accumulate in the intestinal tract until day ten of adulthood. Conclusions: The findings of this study suggest that the nematodes fed the respiratory deficient E. coli diet live longer because the delay in bacterial colonization of the gut subjects the worms to less stress compared to worms fed the OP50 E. coli diet. This work suggests that bacterial respiration can act as a virulence factor, influencing the ability of bacteria to colonize and subsequently harm the animal host. Respiratory deficient bacteria may pose a useful model for probing probiotic relationships within the gut microbiome in higher organisms. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
177. Probucol ameliorates renal and metabolic sequelae of primary CoQ deficiency in Pdss2 mutant mice.
- Author
-
Falk, Marni J., Polyak, Erzsebet, Zhang, Zhe, Peng, Min, King, Rhonda, Maltzman, Jonathan S., Okwuego, Ezinne, Horyn, Oksana, Nakamaru-Ogiso, Eiko, Ostrovsky, Julian, Xie, Letian X., Chen, Jia Yan, Marbois, Beth, Nissim, Itzhak, Clarke, Catherine F., and Gasser, David L.
- Published
- 2011
- Full Text
- View/download PDF
178. Testis-Specific Transcripts of Rat Farnesyl Pyrophosphate Synthetase are Developmentally Regulated and Localized to Haploid Germ Cells1
- Author
-
Teruya, Julie H., primary, Salido, Eduardo C., additional, Edwards, Peter A., additional, and Clarke, Catherine F., additional
- Published
- 1991
- Full Text
- View/download PDF
179. Primary Coenzyme Q Deficiency in Pdss2 Mutant Mice Causes Isolated Renal Disease.
- Author
-
Min Peng, Falk, Marni J., Haase, Volker H., King, Rhonda, Polyak, Erzsebet, Selak, Mary, Yudkoff, Marc, Hancock, Wayne W., Meade, Ray, Saiki, Ryoichi, Lunceford, Adam L., Clarke, Catherine F., and Gasser, David L.
- Subjects
UBIQUINONES ,KIDNEY diseases ,BIOSYNTHESIS ,RESPIRATORY diseases ,LABORATORY mice - Abstract
Coenzyme Q (CoQ) is an essential electron carrier in the respiratory chain whose deficiency has been implicated in a wide variety of human mitochondrial disease manifestations. Its multi-step biosynthesis involves production of polyisoprenoid diphosphate in a reaction that requires the enzymes be encoded by PDSS1 and PDSS2. Homozygous mutations in either of these genes, in humans, lead to severe neuromuscular disease, with nephrotic syndrome seen in PDSS2 deficiency. We now show that a presumed autoimmune kidney disease in mice with the missense Pdss2
kd/kd genotype can be attributed to a mitochondrial CoQ biosynthetic defect. Levels of CoQ9 and CoQ10 in kidney homogenates from B6.Pdss2kd/kd mutants were significantly lower than those in B6 control mice. Disease manifestations originate specifically in glomerular podocytes, as renal disease is seen in Podocin/cre,Pdss2loxP/loxP knockout mice but not in conditional knockouts targeted to renal tubular epithelium, monocytes, or hepatocytes. Liver-conditional B6.Alb/cre,Pdss2loxP/loxP knockout mice have no overt disease despite demonstration that their livers have undetectable CoQ9 levels, impaired respiratory capacity, and significantly altered intermediary metabolism as evidenced by transcriptional profiling and amino acid quantitation. These data suggest that disease manifestations of CoQ deficiency relate to tissue-specific respiratory capacity thresholds, with glomerular podocytes displaying the greatest sensitivity to Pdss2 impairment. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
180. Mitochondrial-ER Contact Sites and Tethers Influence the Biosynthesis and Function of Coenzyme Q.
- Author
-
Novales, Noelle Alexa, Meyer, Hadar, Asraf, Yeynit, Schuldiner, Maya, and Clarke, Catherine F.
- Subjects
- *
UBIQUINONES , *MITOCHONDRIAL membranes , *ELECTRON transport , *ARTIFICIAL membranes , *BIOSYNTHESIS - Abstract
Coenzyme Q (CoQ) is an essential redox-active lipid that plays a major role in the electron transport chain, driving mitochondrial ATP synthesis. In Saccharomyces cerevisiae (yeast), CoQ biosynthesis occurs exclusively in the mitochondrial matrix via a large protein-lipid complex, the CoQ synthome, comprised of CoQ itself, late-stage CoQ-intermediates, and the polypeptides Coq3-Coq9 and Coq11. Coq11 is suggested to act as a negative modulator of CoQ synthome assembly and CoQ synthesis, as its deletion enhances Coq polypeptide content, produces an enlarged CoQ synthome, and restores respiration in mutants lacking the CoQ chaperone polypeptide, Coq10. The CoQ synthome resides in specific niches within the inner mitochondrial membrane, termed CoQ domains, that are often located adjacent to the endoplasmic reticulum-mitochondria encounter structure (ERMES). Loss of ERMES destabilizes the CoQ synthome and renders CoQ biosynthesis less efficient. Here we show that deletion of COQ11 suppresses the respiratory deficient phenotype of select ERMES mutants, results in repair and reorganization of the CoQ synthome, and enhances mitochondrial CoQ domains. Given that ER-mitochondrial contact sites coordinate CoQ biosynthesis, we used a Split-MAM (Mitochondrial Associated Membrane) artificial tether consisting of an ER-mitochondrial contact site reporter, to evaluate the effects of artificial membrane tethers on CoQ biosynthesis in both wild-type and ERMES mutant yeast strains. Overall, this work identifies the deletion of COQ11 as a novel suppressor of phenotypes associated with ERMES deletion mutants and indicates that ER-mitochondria tethers influence CoQ content and turnover, highlighting the role of membrane contact sites in regulating mitochondrial respiratory homeostasis. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
181. Focal segmental glomerulosclerosis is associated with a PDSS2haplotype and, independently, with a decreased content of coenzyme Q10
- Author
-
Gasser, David L., Winkler, Cheryl A., Peng, Min, An, Ping, McKenzie, Louise M., Kirk, Gregory D., Shi, Yuchen, Xie, Letian X., Marbois, Beth N., Clarke, Catherine F., and Kopp, Jeffrey B.
- Abstract
Focal segmental glomerulosclerosis (FSGS) and collapsing glomerulopathy are common causes of nephrotic syndrome. Variants in >20 genes, including genes critical for mitochondrial function, have been associated with these podocyte diseases. One such gene, PDSS2, is required for synthesis of the decaprenyl tail of coenzyme Q10(Q10) in humans. The mouse gene Pdss2is mutated in the kd/kdmouse model of collapsing glomerulopathy. We examined the hypothesis that human PDSS2polymorphisms are associated with podocyte diseases. We genotyped 377 patients with primary FSGS or collapsing glomerulopathy, together with 900 controls, for 9 single-nucleotide polymorphisms in the PDSS2gene in a case-control study. Subjects included 247 African American (AA) and 130 European American (EA) patients and 641 AA and 259 EA controls. Among EAs, a pair of proxy SNPs was significantly associated with podocyte disease, and patients homozygous for one PDSS2haplotype had a strongly increased risk for podocyte disease. By contrast, the distribution of PDSS2genotypes and haplotypes was similar in AA patients and controls. Thus a PDSS2haplotype, which has a frequency of 13% in the EA control population and a homozygote frequency of 1.2%, is associated with a significantly increased risk for FSGS and collapsing glomerulopathy in EAs. Lymphoblastoid cell lines from FSGS patients had significantly less Q10than cell lines from controls; contrary to expectation, this finding was independent of PDSS2haplotype. These results suggest that FSGS patients have Q10deficiency and that this deficiency is manifested in patient-derived lymphoblastoid cell lines.
- Published
- 2013
- Full Text
- View/download PDF
182. Characterization of Saccharomyces cerevisiae ubiquinone-deficient mutants.
- Author
-
Schultz, Jeffery R. and Clarke, Catherine F.
- Subjects
- *
UBIQUINONES , *LINOLENIC acids - Abstract
Abstract. Ubiquinol (QH[sub 2]) is a lipid-soluble molecule that participates in cellular redox reactions. Previous studies have shown that yeast mutants lacking QH[sub 2] are hypersensitive to treatment with polyunsaturated fatty acids (PUFAs) indicating that QH[sub 2] can function as an antioxidant in vivo. In this study the effect of 1 mM linolenic acid on levels of Q[sub 6] and Q[sub 6]H[sub 2] is assessed in both wild-type and respiration-deficient (atp2Delta) strains. The response of Q-deficient mutants to other forms of oxidative stress is further characterized to define those conditions where QH[sub 2] acts as an antioxidant. Endogenous antioxidant defense systems were also assessed in wild-type, Q-deficient, and atp2Delta strains. Superoxide dismutase (SOD) activity decreased and catalase activity increased in both Q-deficient and atp2Delta mutants compared to wild-type cells, suggesting that such changes result from the loss of respiration rather than the lack of Q. [ABSTRACT FROM AUTHOR]
- Published
- 1999
- Full Text
- View/download PDF
183. Coenzyme Q10supplementation rescues renal disease in Pdss2kd/kdmice with mutations in prenyl diphosphate synthase subunit 2
- Author
-
Saiki, Ryoichi, Lunceford, Adam L., Shi, Yuchen, Marbois, Beth, King, Rhonda, Pachuski, Justin, Kawamukai, Makoto, Gasser, David L., and Clarke, Catherine F.
- Abstract
Homozygous mice carrying kd(kidney disease) mutations in the gene encoding prenyl diphosphate synthase subunit 2 (Pdss2kd/kd) develop interstitial nephritis and eventually die from end-stage renal disease. The PDSS2 polypeptide in concert with PDSS1 synthesizes the polyisoprenyl tail of coenzyme Q (Q or ubiquinone), a lipid quinone required for mitochondrial respiratory electron transport. We have shown that a deficiency in Q content is evident in Pdss2kd/kdmouse kidney lipid extracts by 40 days of age and thus precedes the onset of proteinuria and kidney disease by several weeks. The presence of the kd(V117M) mutation in PDSS2 does not prevent its association with PDSS1. However, heterologous expression of the kdmutant form of PDSS2 together with PDSS1 in Escherichia colirecapitulates the Q deficiency observed in the Pdss2kd/kdmouse. Dietary supplementation with Q10provides a dramatic rescue of both proteinuria and interstitial nephritis in the Pdss2kd/kdmutant mice. The results presented suggest that Q may be acting as a potent lipid-soluble antioxidant, rather than by boosting kidney mitochondrial respiration. Such Q10supplementation may have profound and beneficial effects in treatment of certain forms of focal segmental glomerulosclerosis that mirror the renal disease of the Pdss2kd/kdmouse.
- Published
- 2008
- Full Text
- View/download PDF
184. Uptake of Exogenous Coenzyme Q and Transport to Mitochondria Is Required for bc1Complex Stability in Yeast coqMutants*
- Author
-
Santos-Ocaña, Carlos, Do, Thai Q., Clarke, Catherine F., Padilla, Sergio, and Navas, Placido
- Abstract
Coenzyme Q (Q) is an essential component of the mitochondrial respiratory chain in eukaryotic cells but also is present in other cellular membranes where it acts as an antioxidant. Because Q synthesis machinery in Saccharomyces cerevisiaeis located in the mitochondria, the intracellular distribution of Q indicates the existence of intracellular Q transport. In this study, the uptake of exogenous Q6by yeast and its transport from the plasma membrane to mitochondria was assessed in both wild-type and in Q-less coq7mutants derived from four distinct laboratory yeast strains. Q6supplementation of medium containing ethanol, a non-fermentable carbon source, rescued growth in only two of the four coq7mutant strains. Following culture in medium containing dextrose, the added Q6was detected in the plasma membrane of each of four coq7mutants tested. This detection of Q6in the plasma membrane was corroborated by measuring ascorbate stabilization activity, as catalyzed by NADH-ascorbate free radical reductase, a transmembrane redox activity that provides a functional assay of plasma membrane Q6. These assays indicate that each of the four coq7mutant strains assimilate exogenous Q6into the plasma membrane. The two coq7mutant strains rescued by Q6supplementation for growth on ethanol contained mitochondrial Q6levels similar to wild type. However, the content of Q6in mitochondria from the non-rescued strains was only 35 and 8%, respectively, of that present in the corresponding wild-type parental strains. In yeast strains rescued by exogenous Q6, succinate-cytochrome creductase activity was partially restored, whereas non-rescued strains contained very low levels of activity. There was a strong correlation between mitochondrial Q6content, succinate-cytochrome creductase activity, and steady state levels of the cytochrome c1polypeptide. These studies show that transport of extracellular Q6to the mitochondria operates in yeast but is strain-dependent. When Q biosynthesis is disrupted in yeast strains with defects in the intracellular transport of exogenous Q, the bc1complex is unstable. These results indicate that delivery of exogenous Q6to mitochondria is required fore activity and stability of the bc1complex in yeast coqmutants.
- Published
- 2002
- Full Text
- View/download PDF
185. Coenzyme Q Biosynthesis: An Update on the Origins of the Benzenoid Ring and Discovery of New Ring Precursors.
- Author
-
Fernández-del-Río, Lucía and Clarke, Catherine F.
- Subjects
BIOSYNTHESIS ,UBIQUINONES ,STABLE isotopes ,RADIOLABELING ,NATURAL products ,QUINONE ,BENZOQUINONES - Abstract
Coenzyme Q (ubiquinone or CoQ) is a conserved polyprenylated lipid essential for mitochondrial respiration. CoQ is composed of a redox-active benzoquinone ring and a long polyisoprenyl tail that serves as a membrane anchor. A classic pathway leading to CoQ biosynthesis employs 4-hydroxybenzoic acid (4HB). Recent studies with stable isotopes in E. coli, yeast, and plant and animal cells have identified CoQ intermediates and new metabolic pathways that produce 4HB. Stable isotope labeling has identified para-aminobenzoic acid as an alternate ring precursor of yeast CoQ biosynthesis, as well as other natural products, such as kaempferol, that provide ring precursors for CoQ biosynthesis in plants and mammals. In this review, we highlight how stable isotopes can be used to delineate the biosynthetic pathways leading to CoQ. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
186. The mitochondrial carrier SFXN1 is critical for complex III integrity and cellular metabolism.
- Author
-
Acoba, Michelle Grace, Alpergin, Ebru S. Selen, Renuse, Santosh, Fernández-del-Río, Lucía, Lu, Ya-Wen, Khalimonchuk, Oleh, Clarke, Catherine F., Pandey, Akhilesh, Wolfgang, Michael J., and Claypool, Steven M.
- Abstract
Mitochondrial carriers (MCs) mediate the passage of small molecules across the inner mitochondrial membrane (IMM), enabling regulated crosstalk between compartmentalized reactions. Despite MCs representing the largest family of solute carriers in mammals, most have not been subjected to a comprehensive investigation, limiting our understanding of their metabolic contributions. Here, we functionally characterize SFXN1, a member of the non-canonical, sideroflexin family. We find that SFXN1, an integral IMM protein with an uneven number of transmembrane domains, is a TIM22 complex substrate. SFXN1 deficiency leads to mitochondrial respiratory chain impairments, most detrimental to complex III (CIII) biogenesis, activity, and assembly, compromising coenzyme Q levels. The CIII dysfunction is independent of one-carbon metabolism, the known primary role for SFXN1 as a mitochondrial serine transporter. Instead, SFXN1 supports CIII function by participating in heme and α-ketoglutarate metabolism. Our findings highlight the multiple ways that SFXN1-based amino acid transport impacts mitochondrial and cellular metabolic efficiency. [Display omitted] • SFXNs are atypical TIM22 complex substrates • Loss of SFXN1 leads to respiratory chain impairments, most damaging to complex III • SFXN1 contributes to coenzyme Q, heme, α-ketoglutarate, and one-carbon metabolism • Metabolite supplementation improves complex III function in the absence of SFXN1 Acoba et al. show that the amino acid transporter SFXN1 is a cargo of the TIM22 translocon that is important for maintaining complex III function and supports coenzyme Q, heme, and α-ketoglutarate metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
187. Yeast and Rat Coq3 and Escherichia coliUbiG Polypeptides Catalyze Both O-Methyltransferase Steps in Coenzyme Q Biosynthesis*
- Author
-
Poon, Wayne W., Barkovich, Robert J., Hsu, Adam Y., Frankel, Adam, Lee, Peter T., Shepherd, Jennifer N., Myles, David C., and Clarke, Catherine F.
- Abstract
Ubiquinone (coenzyme Q or Q) is a lipid that functions in the electron transport chain in the inner mitochondrial membrane of eukaryotes and the plasma membrane of prokaryotes. Q-deficient mutants of Saccharomyces cerevisiaeharbor defects in one of eight COQgenes (coq1–coq8) and are unable to grow on nonfermentable carbon sources. The biosynthesis of Q involves two separate O-methylation steps. In yeast, the first O-methylation utilizes 3,4-dihydroxy-5-hexaprenylbenzoic acid as a substrate and is thought to be catalyzed by Coq3p, a 32.7-kDa protein that is 40% identical to theEscherichia coli O-methyltransferase, UbiG. In this study, farnesylated analogs corresponding to the secondO-methylation step, demethyl-Q3and Q3, have been chemically synthesized and used to study Q biosynthesis in yeast mitochondria in vitro. Both yeast and rat Coq3p recognize the demethyl-Q3precursor as a substrate. In addition, E. coliUbiGp was purified and found to catalyze both O-methylation steps. Futhermore, antibodies to yeast Coq3p were used to determine that the Coq3 polypeptide is peripherally associated with the matrix-side of the inner membrane of yeast mitochondria. The results indicate that oneO-methyltransferase catalyzes both steps in Q biosynthesis in eukaryotes and prokaryotes and that Q biosynthesis is carried out within the matrix compartment of yeast mitochondria.
- Published
- 1999
- Full Text
- View/download PDF
188. Metabolism of the Flavonol Kaempferol in Kidney Cells Liberates the B-ring to Enter Coenzyme Q Biosynthesis.
- Author
-
Fernández-del-Río, Lucía, Soubeyrand, Eric, Basset, Gilles J., Clarke, Catherine F., Froldi, Guglielmina, and Santos-Ocana, Carlos
- Subjects
BIOSYNTHESIS ,ELECTRON transport ,KIDNEYS ,UBIQUINONES ,CELLS ,METABOLISM - Abstract
Coenzyme Q (CoQ) is an essential component of the mitochondrial electron transport chain and an important antioxidant present in all cellular membranes. CoQ deficiencies are frequent in aging and in age-related diseases, and current treatments are limited to CoQ supplementation. Strategies that rely on CoQ supplementation suffer from poor uptake and trafficking of this very hydrophobic molecule. In a previous study, the dietary flavonol kaempferol was reported to serve as a CoQ ring precursor and to increase the CoQ content in kidney cells, but neither the part of the molecule entering CoQ biosynthesis nor the mechanism were described. In this study, kaempferol labeled specifically in the B-ring was isolated from Arabidopsis plants. Kidney cells treated with this compound incorporated the B-ring of kaempferol into newly synthesized CoQ, suggesting that the B-ring is metabolized via a mechanism described in plant cells. Kaempferol is a natural flavonoid present in fruits and vegetables and possesses antioxidant, anticancer, and anti-inflammatory therapeutic properties. A better understanding of the role of kaempferol as a CoQ ring precursor makes this bioactive compound a potential candidate for the design of interventions aiming to increase endogenous CoQ biosynthesis and may improve CoQ deficient phenotypes in aging and disease. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
189. The COQ7Gene Encodes a Protein in Saccharomyces cerevisiaeNecessary for Ubiquinone Biosynthesis (∗)
- Author
-
Marbois, B. Noelle and Clarke, Catherine F.
- Abstract
Ubiquinone (coenzyme Q) is a lipid that transports electrons in the respiratory chains of both prokaryotes and eukaryotes. Mutants of Saccharomyces cerevisiaedeficient in ubiquinone biosynthesis fail to grow on nonfermentable carbon sources and have been classified into eight complementation groups (coq1-coq8; Tzagoloff, A., and Dieckmann, C. L.(1990) Microbiol. Rev.54, 211-225). In this study we show that although yeast coq7mutants lack detectable ubiquinone, the coq7-1mutant does synthesize demethoxyubiquinone (2-hexaprenyl-3-methyl-6-methoxy-1,4-benzoquinone), a ubiquinone biosynthetic intermediate. The corresponding wild-type COQ7gene was isolated, sequenced, and found to restore growth on nonfermentable carbon sources and the synthesis of ubiquinone. The sequence predicts a polypeptide of 272 amino acids which is 40% identical to a previously reported Caenorhabditis elegansopen reading frame. Deletion of the chromosomal COQ7gene generates respiration defective yeast mutants deficient in ubiquinone. Analysis of several coq7deletion strains indicates that, unlike the coq7-1mutant, demethoxyubiquinone is not produced. Both coq7-1and coq7deletion mutants, like other coqmutants, accumulate an early intermediate in the ubiquinone biosynthetic pathway, 3-hexaprenyl-4-hydroxybenzoate. The data suggest that the yeast COQ7gene may encode a protein involved in one or more monoxygenase or hydroxylase steps of ubiquinone biosynthesis.
- Published
- 1996
- Full Text
- View/download PDF
190. Nonfunctional coq10 mutants maintain the ERMES complex and reveal true phenotypes associated with the loss of the coenzyme Q chaperone protein Coq10.
- Author
-
Novales, Noelle Alexa, Feustel, Kelsey J., He, Kevin L., Chanfreau, Guillaume F., and Clarke, Catherine F.
- Subjects
- *
MOLECULAR chaperones , *ELECTRON transport , *EXTRACELLULAR matrix proteins , *SACCHAROMYCES cerevisiae , *BIOSYNTHESIS - Abstract
Coenzyme Q (CoQ) is a redox-active lipid molecule that acts as an electron carrier in the mitochondrial electron transport chain. In Saccharomyces cerevisiae, CoQ is synthesized in the mitochondrial matrix by a multisubunit protein--lipid complex termed the CoQ synthome, the spatial positioning of which is coordinated by the endoplasmic reticulum-mitochondria encounter structure (ERMES). The MDM12 gene encoding the cytosolic subunit of ERMES is coexpressed with COQ10, which encodes the putative CoQ chaperone Coq10, via a shared bidirectional promoter. Deletion of COQ10 results in respiratory deficiency, impaired CoQ biosynthesis, and reduced spatial coordination between ERMES and the CoQ synthome. While Coq10 protein content is maintained upon deletion of MDM12, we show that deletion of COQ10 by replacement with a HIS3 marker results in diminished Mdm12 protein content. Since deletion of individual ERMES subunits prevents ERMES formation, we asked whether some or all of the phenotypes associated with COQ10 deletion result from ERMES dysfunction. To identify the phenotypes resulting solely due to the loss of Coq10, we constructed strains expressing a functionally impaired (coq10-L96S) or truncated (coq10-R147*) Coq10 isoform using CRISPR-Cas9. We show that both coq10 mutants preserve Mdm12 protein content and exhibit impaired respiratory capacity like the coq10Δ mutant, indicating that Coq10's function is vital for respiration regardless of ERMES integrity. Moreover, the maintenance of CoQ synthome stability and efficient CoQ biosynthesis observed for the coq10-R147* mutant suggests these deleterious phenotypes in the coq10Δ mutant result from ERMES disruption. Overall, this study clarifies the role of Coq10 in modulating CoQ biosynthesis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
191. Genetic screening reveals phospholipid metabolism as a key regulator of the biosynthesis of the redox-active lipid coenzyme Q
- Author
-
Ayer, Anita, Fazakerley, Daniel J, Suarna, Cacang, Maghzal, Ghassan J, Sheipouri, Diba, Lee, Kevin J, Bradley, Michelle C, Fernández-Del-Rio, Lucía, Tumanov, Sergey, Kong, Stephanie My, Van Der Veen, Jelske N, Yang, Andrian, Ho, Joshua WK, Clarke, Steven G, James, David E, Dawes, Ian W, Vance, Dennis E, Clarke, Catherine F, Jacobs, René L, and Stocker, Roland
- Subjects
S-adenosylmethionine ,Mitochondrial Diseases ,Ubiquinone ,Phosphatidylethanolamine N-Methyltransferase ,S-adenosylhomocysteine ,food and beverages ,Coenzyme Q ,Insulin resistance ,3. Good health ,Mitochondria ,Mice ,PEMT ,Animals ,Genetic Testing ,Reactive oxygen species ,Oxidation-Reduction ,Phospholipids - Abstract
Mitochondrial energy production and function rely on optimal concentrations of the essential redox-active lipid, coenzyme Q (CoQ). CoQ deficiency results in mitochondrial dysfunction associated with increased mitochondrial oxidative stress and a range of pathologies. What drives CoQ deficiency in many of these pathologies is unknown, just as there currently is no effective therapeutic strategy to overcome CoQ deficiency in humans. To date, large-scale studies aimed at systematically interrogating endogenous systems that control CoQ biosynthesis and their potential utility to treat disease have not been carried out. Therefore, we developed a quantitative high-throughput method to determine CoQ concentrations in yeast cells. Applying this method to the Yeast Deletion Collection as a genome-wide screen, 30 genes not known previously to regulate cellular concentrations of CoQ were discovered. In combination with untargeted lipidomics and metabolomics, phosphatidylethanolamine N-methyltransferase (PEMT) deficiency was confirmed as a positive regulator of CoQ synthesis, the first identified to date. Mechanistically, PEMT deficiency alters mitochondrial concentrations of one-carbon metabolites, characterized by an increase in the S-adenosylmethionine to S-adenosylhomocysteine (SAM-to-SAH) ratio that reflects mitochondrial methylation capacity, drives CoQ synthesis, and is associated with a decrease in mitochondrial oxidative stress. The newly described regulatory pathway appears evolutionary conserved, as ablation of PEMT using antisense oligonucleotides increases mitochondrial CoQ in mouse-derived adipocytes that translates to improved glucose utilization by these cells, and protection of mice from high-fat diet-induced insulin resistance. Our studies reveal a previously unrecognized relationship between two spatially distinct lipid pathways with potential implications for the treatment of CoQ deficiencies, mitochondrial oxidative stress/dysfunction, and associated diseases.
192. Genetic screening reveals phospholipid metabolism as a key regulator of the biosynthesis of the redox-active lipid coenzyme Q
- Author
-
Ayer, Anita, Fazakerley, Daniel J, Suarna, Cacang, Maghzal, Ghassan J, Sheipouri, Diba, Lee, Kevin J, Bradley, Michelle C, Fernández-Del-Rio, Lucía, Tumanov, Sergey, Kong, Stephanie My, Van Der Veen, Jelske N, Yang, Andrian, Ho, Joshua W K, Clarke, Steven G, James, David E, Dawes, Ian W, Vance, Dennis E, Clarke, Catherine F, Jacobs, René L, and Stocker, Roland
- Subjects
S-Adenosylmethionine ,Pemt ,S-adenosylhomocysteine ,food and beverages ,Coenzyme Q ,Insulin resistance ,Reactive oxygen species ,3. Good health ,Mitochondria - Abstract
Mitochondrial energy production and function rely on optimal concentrations of the essential redox-active lipid, coenzyme Q (CoQ). CoQ deficiency results in mitochondrial dysfunction associated with increased mitochondrial oxidative stress and a range of pathologies. What drives CoQ deficiency in many of these pathologies is unknown, just as there currently is no effective therapeutic strategy to overcome CoQ deficiency in humans. To date, large-scale studies aimed at systematically interrogating endogenous systems that control CoQ biosynthesis and their potential utility to treat disease have not been carried out. Therefore, we developed a quantitative high-throughput method to determine CoQ concentrations in yeast cells. Applying this method to the Yeast Deletion Collection as a genome-wide screen, 30 genes not known previously to regulate cellular concentrations of CoQ were discovered. In combination with untargeted lipidomics and metabolomics, phosphatidylethanolamine N-methyltransferase (PEMT) deficiency was confirmed as a positive regulator of CoQ synthesis, the first identified to date. Mechanistically, PEMT deficiency alters mitochondrial concentrations of one-carbon metabolites, characterized by an increase in the S-adenosylmethionine to S-adenosylhomocysteine (SAM-to-SAH) ratio that reflects mitochondrial methylation capacity, drives CoQ synthesis, and is associated with a decrease in mitochondrial oxidative stress. The newly described regulatory pathway appears evolutionary conserved, as ablation of PEMT using antisense oligonucleotides increases mitochondrial CoQ in mouse-derived adipocytes that translates to improved glucose utilization by these cells, and protection of mice from high-fat diet-induced insulin resistance. Our studies reveal a previously unrecognized relationship between two spatially distinct lipid pathways with potential implications for the treatment of CoQ deficiencies, mitochondrial oxidative stress/dysfunction, and associated diseases.
193. Osmotic stress: Is CoQ a membrane stabilizer?
- Author
-
Clarke, Catherine F, Rowat, Amy C, and Gober, James W
- Subjects
- *
BILAYER lipid membranes , *PHYSIOLOGY , *UBIQUINONES , *OSMOTIC pressure , *MOLECULAR structure of cellular membranes , *ISOPRENE - Abstract
The article focuses on a study which shows that lipid membranes that contain coenzyme Q (CoQ) in bacterial species Escheria Coli with less than eight isoprene units may have enhanced mechanical stabilization. According to the study, bacterial tolerance of salt stress is mediated by flux control of water across the cell membrane, adjustments of intracellular potassium levels and transport of osmoproctents.
- Published
- 2014
- Full Text
- View/download PDF
194. Carbonyl Posttranslational Modification Associated With Early-Onset Type 1 Diabetes Autoimmunity.
- Author
-
Yang, Mei-Ling, Connolly, Sean E., Gee, Renelle J., Lam, TuKiet T., Kanyo, Jean, Peng, Jian, Guyer, Perrin, Syed, Farooq, Tse, Hubert M., Clarke, Steven G., Clarke, Catherine F., James, Eddie A., Speake, Cate, Evans-Molina, Carmella, Arvan, Peter, Herold, Kevan C., Wen, Li, and Mamula, Mark J.
- Subjects
- *
PROTEIN metabolism , *PROTEINS , *ANIMAL experimentation , *TYPE 1 diabetes , *ISLANDS of Langerhans , *INSULIN , *PROINSULIN , *IMMUNITY , *RESEARCH funding , *MICE , *ANTIGENS - Abstract
Inflammation and oxidative stress in pancreatic islets amplify the appearance of various posttranslational modifications to self-proteins. In this study, we identified a select group of carbonylated islet proteins arising before the onset of hyperglycemia in NOD mice. Of interest, we identified carbonyl modification of the prolyl-4-hydroxylase β subunit (P4Hb) that is responsible for proinsulin folding and trafficking as an autoantigen in both human and murine type 1 diabetes. We found that carbonylated P4Hb is amplified in stressed islets coincident with decreased glucose-stimulated insulin secretion and altered proinsulin-to-insulin ratios. Autoantibodies against P4Hb were detected in prediabetic NOD mice and in early human type 1 diabetes prior to the onset of anti-insulin autoimmunity. Moreover, we identify autoreactive CD4+ T-cell responses toward carbonyl-P4Hb epitopes in the circulation of patients with type 1 diabetes. Our studies provide mechanistic insight into the pathways of proinsulin metabolism and in creating autoantigenic forms of insulin in type 1 diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
195. A dedicated flavin-dependent monooxygenase catalyzes the hydroxylation of demethoxyubiquinone into ubiquinone (coenzyme Q) in Arabidopsis.
- Author
-
Latimer, Scott, Keene, Shea A., Stutts, Lauren R., Berger, Antoine, Bernert, Ann C., Soubeyrand, Eric, Wright, Janet, Clarke, Catherine F., Block, Anna K., Colquhoun, Thomas A., Elowsky, Christian, Christensen, Alan, Wilson, Mark A., and Basset, Gilles J.
- Subjects
- *
UBIQUINONES , *MONOOXYGENASES , *HORIZONTAL gene transfer , *HYDROXYLATION , *ARABIDOPSIS , *GENE regulatory networks - Abstract
Ubiquinone (Coenzyme Q) is a vital respiratory cofactor and liposoluble antioxidant. In plants, it is not known how the C-6 hydroxylation of demethoxyubiquinone, the penultimate step in ubiquinone biosynthesis, is catalyzed. The combination of cross-species gene network modeling along with mining of embryo-defective mutant databases of Arabidopsis thaliana identified the embryo lethal locus EMB2421 (At1g24340) as a top candidate for the missing plant demethoxyubiquinone hydroxylase. In marked contrast with prototypical eukaryotic demethoxyubiquinone hydroxylases, the catalytic mechanism of which depends on a carboxylate-bridged di-iron domain, At1g24340 is homologous to FAD-dependent oxidoreductases that instead use NAD(P)H as an electron donor. Complementation assays in Saccharomyces cerevisiae and Escherichia coli demonstrated that At1g24340 encodes a functional demethoxyubiquinone hydroxylase and that the enzyme displays strict specificity for the C-6 position of the benzoquinone ring. Laser-scanning confocal microscopy also showed that GFP-tagged At1g24340 is targeted to mitochondria. Silencing of At1g24340 resulted in 40 to 74% decrease in ubiquinone content and de novo ubiquinone biosynthesis. Consistent with the role of At1g24340 as a benzenoid ring modification enzyme, this metabolic blockage could not be bypassed by supplementation with 4-hydroxybenzoate, the immediate precursor of ubiquinone's ring. Unlike in yeast, in Arabidopsis overexpression of demethoxyubiquinone hydroxylase did not boost ubiquinone content. Phylogenetic reconstructions indicated that plant demethoxyubiquinone hydroxylase is most closely related to prokaryotic monooxygenases that act on halogenated aromatics and likely descends from an event of horizontal gene transfer between a green alga and a bacterium. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
196. Intragenic suppressor mutations of the COQ8 protein kinase homolog restore coenzyme Q biosynthesis and function in Saccharomyces cerevisiae.
- Author
-
Awad, Agape M., Nag, Anish, Pham, Nguyen V. B., Bradley, Michelle C., Jabassini, Nour, Nathaniel, Juan, and Clarke, Catherine F.
- Subjects
- *
SUPPRESSOR mutation , *PROTEIN kinases , *SACCHAROMYCES cerevisiae , *BIOSYNTHESIS , *UBIQUINONES - Abstract
Saccharomyces cerevisiae Coq8 is a member of the ancient UbiB atypical protein kinase family. Coq8, and its orthologs UbiB, ABC1, ADCK3, and ADCK4, are required for the biosynthesis of coenzyme Q in yeast, E. coli, A. thaliana, and humans. Each Coq8 ortholog retains nine highly conserved protein kinase-like motifs, yet its functional role in coenzyme Q biosynthesis remains mysterious. Coq8 may function as an ATPase whose activity is stimulated by coenzyme Q intermediates and phospholipids. A key yeast point mutant expressing Coq8-A197V was previously shown to result in a coenzyme Q-less, respiratory deficient phenotype. The A197V substitution occurs in the crucial Ala-rich protein kinase-like motif I of yeast Coq8. Here we show that long-term cultures of mutants expressing Coq8-A197V produce spontaneous revertants with the ability to grow on medium containing a non-fermentable carbon source. Each revertant is shown to harbor a secondary intragenic suppressor mutation within the COQ8 gene. The intragenic suppressors restore the synthesis of coenzyme Q. One class of the suppressors fully restores the levels of coenzyme Q and key Coq polypeptides necessary for the maintenance and integrity of the high-molecular mass CoQ synthome (also termed complex Q), while the other class provides only a partial rescue. Mutants harboring the first class of suppressors grow robustly under respiratory conditions, while mutants containing the second class grow more slowly under these conditions. Our work provides insight into the function of this important yet still enigmatic Coq8 family. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
197. COQ11 deletion mitigates respiratory deficiency caused by mutations in the gene encoding the coenzyme Q chaperone protein Coq10.
- Author
-
Bradley, Michelle C., Yang, Krista, Fernández-del-Río, Lucía, Ngo, Jennifer, Ayer, Anita, Tsui, Hui S., Novales, Noelle Alexa, Stocker, Roland, Shirihai, Orian S., Barros, Mario H., and Clarke, Catherine F.
- Subjects
- *
GENETIC mutation , *UBIQUINONES , *ELECTRON transport , *OXIDATIVE phosphorylation , *MOLECULAR chaperones , *BIOSYNTHESIS , *ENERGY metabolism , *GROWTH plate - Abstract
Coenzyme Q (Qn) is a vital lipid component of the electron transport chain that functions in cellular energy metabolism and as a membrane antioxidant. In the yeast Saccharomyces cerevisiae, coq1-coq9 deletion mutants are respiratory-incompetent, sensitive to lipid peroxidation stress, and unable to synthesize Q6. The yeast coq10 deletion mutant is also respiratorydeficient and sensitive to lipid peroxidation, yet it continues to produce Q6 at an impaired rate. Thus, Coq10 is required for the function of Q6 in respiration and as an antioxidant and is believed to chaperone Q6 from its site of synthesis to the respiratory complexes. In several fungi, Coq10 is encoded as a fusion polypeptide with Coq11, a recently identified protein of unknown function required for efficient Q6 biosynthesis. Because "fused" proteins are often involved in similar biochemical pathways, here we examined the putative functional relationship between Coq10 and Coq11 in yeast. We used plate growth and Seahorse assays and LC-MS/MS analysis to show that COQ11 deletion rescues respiratory deficiency, sensitivity to lipid peroxidation, and decreased Q6 biosynthesis of the coq10Δ mutant. Additionally, immunoblotting indicated that yeast coq11Δ mutants accumulate increased amounts of certain Coq polypeptides and display a stabilized CoQ synthome. These effects suggest that Coq11 modulates Q6 biosynthesis and that its absence increases mitochondrial Q6 content in the coq10Δcoq11Δ double mutant. This augmented mitochondrial Q6 content counteracts the respiratory deficiency and lipid peroxidation sensitivity phenotypes of the coq10Δ mutant. This study further clarifies the intricate connection between Q6 biosynthesis, trafficking, and function in mitochondrial metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
198. Recombinant RquA catalyzes the in vivo conversion of ubiquinone to rhodoquinone in Escherichia coli and Saccharomyces cerevisiae.
- Author
-
Bernert, Ann C., Jacobs, Evan J., Reinl, Samantha R., Choi, Christina C.Y., Roberts Buceta, Paloma M., Culver, John C., Goodspeed, Carly R., Bradley, Michelle C., Clarke, Catherine F., Basset, Gilles J., and Shepherd, Jennifer N.
- Subjects
- *
SACCHAROMYCES cerevisiae , *ESCHERICHIA coli , *QUINONE , *REVERSE genetics , *CHARGE exchange , *ELECTROPHILES - Abstract
Terpenoid quinones are liposoluble redox-active compounds that serve as essential electron carriers and antioxidants. One such quinone, rhodoquinone (RQ), couples the respiratory electron transfer chain to the reduction of fumarate to facilitate anaerobic respiration. This mechanism allows RQ-synthesizing organisms to operate their respiratory chain using fumarate as a final electron acceptor. RQ biosynthesis is restricted to a handful of prokaryotic and eukaryotic organisms, and details of this biosynthetic pathway remain enigmatic. One gene, rquA , was discovered to be required for RQ biosynthesis in Rhodospirillum rubrum. However, the function of the gene product, RquA, has remained unclear. Here, using reverse genetics approaches, we demonstrate that RquA converts ubiquinone to RQ directly. We also demonstrate the first in vivo synthetic production of RQ in Escherichia coli and Saccharomyces cerevisiae , two organisms that do not natively produce RQ. These findings help clarify the complete RQ biosynthetic pathway in species which contain RquA homologs. • A complete pathway for rhodoquinone biosynthesis in rquA -producing species has been elucidated. • Recombinant RquA is active in two non rhodoquinone-producing species, Escherichia coli and Saccharomyces cerevisiae. • Expression of RquA in E. coli and S. cerevisiae facilitates the in vivo synthesis of rhodoquinone. • Ubiquinone is a required substrate of RquA, and the product of the RquA reaction is rhodoquinone. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
199. Chromatin-remodeling SWI/SNF complex regulates coenzyme Q6 synthesis and a metabolic shift to respiration in yeast.
- Author
-
Awad, Agape M., Venkataramanan, Srivats, Nag, Anish, Galivanche, Anoop Raj, Bradley, Michelle C., Neves, Lauren T., Douglass, Stephen, Clarke, Catherine F., and Johnson, Tracy L.
- Subjects
- *
CHROMATIN-remodeling complexes , *CHROMATIN , *CELLULAR control mechanisms , *RESPIRATION , *PHYSIOLOGY ,YEAST physiology - Abstract
Despite its relatively streamlined genome, there are many important examples of regulated RNA splicing in Saccharomyces cerevisiae. Here, we report a role for the chromatin remodeler SWI/SNF in respiration, partially via the regulation of splicing. Wefind that a nutrient-dependent decrease in Snf2 leads to an increase in splicing of the PTC7 transcript. The spliced PTC7 transcript encodes a mitochondrial phosphatase regulator of biosynthesis of coenzyme Q6 (ubiquinone or CoQ6) and a mitochondrial redox-active lipid essential for electron and proton transport in respiration. Increased splicing of PTC7 increases CoQ6 levels. The increase in PTC7 splicing occurs at least in part due to down-regulation of ribosomal protein gene expression, leading to the redistribution of spliceosomes from this abundant class of intron-containing RNAs to otherwise poorly spliced transcripts. In contrast, a protein encoded by the nonspliced isoform of PTC7 represses CoQ6 biosynthesis. Taken together, these findings uncover a link between Snf2 expression and the splicing of PTC7 and establish a previously unknown role for the SWI/SNF complex in the transition of yeast cells from fermentative to respiratory modes of metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
200. Yeast Coq9 controls deamination of coenzyme Q intermediates that derive from para-aminobenzoic acid.
- Author
-
He, Cuiwen H., Black, Dylan S., Nguyen, Theresa P.T., Wang, Charles, Srinivasan, Chandra, and Clarke, Catherine F.
- Subjects
- *
POLYPEPTIDES , *YEAST , *UBIQUINONES , *PARA-aminobenzoic acid , *GENETIC overexpression , *BIOSYNTHESIS - Abstract
Coq9 is a polypeptide subunit in a mitochondrial multi-subunit complex, termed the CoQ-synthome, required for biosynthesis of coenzyme Q (ubiquinone or Q). Deletion of COQ9 results in dissociation of the CoQ-synthome, but over-expression of Coq8 putative kinase stabilizes the CoQ-synthome in the coq9 null mutant and leads to the accumulation of two nitrogen-containing Q intermediates, imino-demethoxy-Q 6 (IDMQ 6 ) and 3-hexaprenyl-4-aminophenol (4-AP) when para -aminobenzoic acid (pABA) is provided as a ring precursor. To investigate whether Coq9 is responsible for deamination steps in Q biosynthesis, we utilized the yeast coq5-5 point mutant. The yeast coq5-5 point mutant is defective in the C -methyltransferase step of Q biosynthesis but retains normal steady-state levels of the Coq5 polypeptide. Here, we show that when high amounts of 13 C 6 -pABA are provided, the coq5-5 mutant accumulates both 13 C 6 -imino-demethyl-demethoxy-Q 6 ( 13 C 6 -IDDMQ 6 ) and 13 C 6 -demethyl-demethoxy-Q 6 ( 13 C 6 -DDMQ 6 ). Deletion of COQ9 in the yeast coq5-5 mutant along with Coq8 over-expression and 13 C 6 - pABA labeling leads to the absence of 13 C 6 -DDMQ 6 , and the nitrogen-containing intermediates 13 C 6 -4-AP and 13 C 6 -IDDMQ 6 persist. We describe a coq9 temperature-sensitive mutant and show that at the non-permissive temperature, steady-state polypeptide levels of Coq9-ts19 increased, while Coq4, Coq5, Coq6, and Coq7 decreased. The coq9-ts19 mutant had decreased Q 6 content and increased levels of nitrogen-containing intermediates. These findings identify Coq9 as a multi-functional protein that is required for the function of Coq6 and Coq7 hydroxylases, for removal of the nitrogen substituent from pABA-derived Q intermediates, and is an essential component of the CoQ synthome. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.