117 results on '"Catarina M. Quinzii"'
Search Results
2. Mechanisms and Therapeutic Effects of Benzoquinone Ring Analogs in Primary CoQ Deficiencies
- Author
-
Alba Pesini, Agustin Hidalgo-Gutierrez, and Catarina M. Quinzii
- Subjects
coenzyme Q10 ,analogs ,4-hydroxybenzoic acid ,mitochondria ,preclinical models ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Coenzyme Q (CoQ) is a conserved polyprenylated lipid composed of a redox-active benzoquinone ring and a long polyisoprenyl tail that serves as a membrane anchor. CoQ biosynthesis involves multiple steps, including multiple modifications of the precursor ring 4-hydroxybenzoic acid. Mutations in the enzymes involved in CoQ biosynthesis pathway result in primary coenzyme Q deficiencies, mitochondrial disorders whose clinical heterogenicity reflects the multiple biological function of CoQ. Patients with these disorders do not always respond to CoQ supplementation, and CoQ analogs have not been successful as alternative approaches. Progress made in understanding the CoQ biosynthesis pathway and studies of supplementation with 4-hydroxybenzoic acid ring analogs have opened a new area in the field of primary CoQ deficiencies treatment. Here, we will review these studies, focusing on efficacy of the different 4-hydroxybenzoic acid ring analogs, models in which they have been tested, and their mechanisms of action. Understanding how these compounds ameliorate biochemical, molecular, and/or clinical phenotypes of CoQ deficiencies is important to develop the most rational treatment for CoQ deficient patients, depending on their molecular defects.
- Published
- 2022
- Full Text
- View/download PDF
3. Mitochondrial Dysregulation and Impaired Autophagy in iPSC-Derived Dopaminergic Neurons of Multiple System Atrophy
- Author
-
Giacomo Monzio Compagnoni, Giulio Kleiner, Maura Samarani, Massimo Aureli, Gaia Faustini, Arianna Bellucci, Dario Ronchi, Andreina Bordoni, Manuela Garbellini, Sabrina Salani, Francesco Fortunato, Emanuele Frattini, Elena Abati, Christian Bergamini, Romana Fato, Silvia Tabano, Monica Miozzo, Giulia Serratto, Maria Passafaro, Michela Deleidi, Rosamaria Silipigni, Monica Nizzardo, Nereo Bresolin, Giacomo P. Comi, Stefania Corti, Catarina M. Quinzii, and Alessio Di Fonzo
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Multiple system atrophy (MSA) is a progressive neurodegenerative disease that affects several areas of the CNS, whose pathogenesis is still widely unclear and for which an effective treatment is lacking. We have generated induced pluripotent stem cell-derived dopaminergic neurons from four MSA patients and four healthy controls and from two monozygotic twins discordant for the disease. In this model, we have demonstrated an aberrant autophagic flow and a mitochondrial dysregulation involving respiratory chain activity, mitochondrial content, and CoQ10 biosynthesis. These defective mechanisms may contribute to the onset of the disease, representing potential therapeutic targets. : Monzio Compagnoni et al. present an iPSC-based neuronal in vitro model of multiple system atrophy. Patients' dopaminergic neurons display a dysregulation of mitochondrial functioning and autophagy, suggesting new hints for the comprehension of the pathogenesis of the disease. Keywords: multiple system atrophy, induced pluripotent stem cells, dopaminergic neurons, mitochondria, autophagy, MSA, neurodegeneration
- Published
- 2018
- Full Text
- View/download PDF
4. Inhibition of Bcl-2/Bcl-xL and c-MET causes synthetic lethality in model systems of glioblastoma
- Author
-
Yiru Zhang, Chiaki Tsuge Ishida, Chang Shu, Giulio Kleiner, Maria J. Sanchez-Quintero, Elena Bianchetti, Catarina M. Quinzii, Mike-Andrew Westhoff, Georg Karpel-Massler, and Markus D. Siegelin
- Subjects
Medicine ,Science - Abstract
Abstract Recent data suggest that glioblastomas (GBM) activate the c-MET signaling pathway and display increased levels in anti-apoptotic Bcl-2 family members. Therefore, targeting these two deregulated pathways for therapy might yield synergistic treatment responses. We applied extracellular flux analysis to assess tumor metabolism. We found that combined treatment with ABT263 and Crizotinib synergistically reduces the proliferation of glioblastoma cells, which was dependent on dual inhibition of Bcl-2 and Bcl-xL. The combination treatment led to enhanced apoptosis with loss of mitochondrial membrane potential and activation of caspases. On the molecular level, c-MET-inhibition results in significant energy deprivation with a reduction in oxidative phosphorylation, respiratory capacity and a suppression of intracellular energy production (ATP). In turn, loss of energy levels suppresses protein synthesis, causing a decline in anti-apoptotic Mcl-1 levels. Silencing of Mcl-1 enhanced ABT263 and MET-inhibitor mediated apoptosis, but marginally the combination treatment, indicating that Mcl-1 is the central factor for the induction of cell death induced by the combination treatment. Finally, combined treatment with BH3-mimetics and c-MET inhibitors results in significantly smaller tumors than each treatment alone in a PDX model system of glioblastoma. These results suggest that c-MET inhibition causes a selective vulnerability of GBM cells to Bcl-2/Bcl-xL inhibition.
- Published
- 2018
- Full Text
- View/download PDF
5. Metabolic Targets of Coenzyme Q10 in Mitochondria
- Author
-
Agustín Hidalgo-Gutiérrez, Pilar González-García, María Elena Díaz-Casado, Eliana Barriocanal-Casado, Sergio López-Herrador, Catarina M. Quinzii, and Luis C. López
- Subjects
coenzyme Q10 ,ubiquinone-10 ,ubiquinol-10 ,mitochondria ,OxPhos ,super-complexes ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Coenzyme Q10 (CoQ10) is classically viewed as an important endogenous antioxidant and key component of the mitochondrial respiratory chain. For this second function, CoQ molecules seem to be dynamically segmented in a pool attached and engulfed by the super-complexes I + III, and a free pool available for complex II or any other mitochondrial enzyme that uses CoQ as a cofactor. This CoQ-free pool is, therefore, used by enzymes that link the mitochondrial respiratory chain to other pathways, such as the pyrimidine de novo biosynthesis, fatty acid β-oxidation and amino acid catabolism, glycine metabolism, proline, glyoxylate and arginine metabolism, and sulfide oxidation metabolism. Some of these mitochondrial pathways are also connected to metabolic pathways in other compartments of the cell and, consequently, CoQ could indirectly modulate metabolic pathways located outside the mitochondria. Thus, we review the most relevant findings in all these metabolic functions of CoQ and their relations with the pathomechanisms of some metabolic diseases, highlighting some future perspectives and potential therapeutic implications.
- Published
- 2021
- Full Text
- View/download PDF
6. Inhibition of HDAC1/2 Along with TRAP1 Causes Synthetic Lethality in Glioblastoma Model Systems
- Author
-
Trang T. T. Nguyen, Yiru Zhang, Enyuan Shang, Chang Shu, Catarina M. Quinzii, Mike-Andrew Westhoff, Georg Karpel-Massler, and Markus D. Siegelin
- Subjects
glioblastoma ,gamitrinib ,HDAC inhibitors ,tumor metabolism ,electron transport chain ,Bcl-2 family ,Cytology ,QH573-671 - Abstract
The heterogeneity of glioblastomas, the most common primary malignant brain tumor, remains a significant challenge for the treatment of these devastating tumors. Therefore, novel combination treatments are warranted. Here, we showed that the combined inhibition of TRAP1 by gamitrinib and histone deacetylases (HDAC1/HDAC2) through romidepsin or panobinostat caused synergistic growth reduction of established and patient-derived xenograft (PDX) glioblastoma cells. This was accompanied by enhanced cell death with features of apoptosis and activation of caspases. The combination treatment modulated the levels of pro- and anti-apoptotic Bcl-2 family members, including BIM and Noxa, Mcl-1, Bcl-2 and Bcl-xL. Silencing of Noxa, BAK and BAX attenuated the effects of the combination treatment. At the metabolic level, the combination treatment led to an enhanced reduction of oxygen consumption rate and elicited an unfolded stress response. Finally, we tested whether the combination treatment of gamitrinib and panobinostat exerted therapeutic efficacy in PDX models of glioblastoma (GBM) in mice. While single treatments led to mild to moderate reduction in tumor growth, the combination treatment suppressed tumor growth significantly stronger than single treatments without induction of toxicity. Taken together, we have provided evidence that simultaneous targeting of TRAP1 and HDAC1/2 is efficacious to reduce tumor growth in model systems of glioblastoma.
- Published
- 2020
- Full Text
- View/download PDF
7. The Role of Sulfide Oxidation Impairment in the Pathogenesis of Primary CoQ Deficiency
- Author
-
Catarina M. Quinzii, Marta Luna-Sanchez, Marcello Ziosi, Agustin Hidalgo-Gutierrez, Giulio Kleiner, and Luis C. Lopez
- Subjects
coenzyme Q ,CoQ ,sulfides ,H2S ,sulfide:quinone oxidoreductase ,SQOR ,Physiology ,QP1-981 - Abstract
Coenzyme Q (CoQ) is a lipid present in all cell membranes. One of the multiple metabolic functions of CoQ is to transport electrons in the reaction catalyzed by sulfide:quinone oxidoreductase (SQOR), the first enzyme of the oxidation pathway of sulfides (hydrogen sulfide, H2S). Early evidence of a defect in the metabolism of H2S in primary CoQ deficiency came from yeast studies in Schizosaccharomyces pombe strains defective for dps1 and ppt1 (homologs of PDSS1 and COQ2, respectively), which have H2S accumulation. Our recent studies in human skin fibroblasts and in murine models of primary CoQ deficiency show that, also in mammals, decreased CoQ levels cause impairment of H2S oxidation. Patient fibroblasts carrying different mutations in genes encoding proteins involved in CoQ biosynthesis show reduced SQOR activity and protein levels proportional to the levels of CoQ. In Pdss2kd/kd mice, kidney, the only organ clinically affected, shows reduced SQOR levels and downstream enzymes, accumulation of H2S, and glutathione depletion. Pdss2kd/kd mice have also low levels of thiosulfate in plasma and urine, and increased C4–C6 acylcarnitines in blood, due to inhibition of short-chain acyl-CoA dehydrogenase. Also in Coq9R239X mice, the symptomatic organ, cerebrum, shows accumulation of H2S, reduced SQOR, increase in thiosulfate sulfurtransferase and sulfite oxidase, and reduction in the levels of glutathione and glutathione enzymes, leading to alteration of the biosynthetic pathways of glutamate, serotonin, and catecholamines. Coq9R239X mice have also reduced blood pressure, possible consequence of H2S-induced vasorelaxation. Since liver is not clinically affected in Pdss2 and Coq9 mutant mice, the effects of the impairment of H2S oxidation in this organ were not investigated, despite its critical role in metabolism. In conclusion, in vitro and in vivo studies of CoQ deficient models provide evidence of tissue-specific H2S oxidation impairment, an additional pathomechanism that should be considered in the understanding and treatment of primary CoQ deficiency.
- Published
- 2017
- Full Text
- View/download PDF
8. Activation of LXRβ inhibits tumor respiration and is synthetically lethal with Bcl‐xL inhibition
- Author
-
Trang Thi Thu Nguyen, Chiaki Tsuge Ishida, Enyuan Shang, Chang Shu, Consuelo Torrini, Yiru Zhang, Elena Bianchetti, Maria J Sanchez‐Quintero, Giulio Kleiner, Catarina M Quinzii, Mike‐Andrew Westhoff, Georg Karpel‐Massler, Peter Canoll, and Markus D Siegelin
- Subjects
BH3 mimetics ,colon adenocarcinoma ,electron transport chain ,glioblastoma ,LXR agonist ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Liver‐X‐receptor (LXR) agonists are known to bear anti‐tumor activity. However, their efficacy is limited and additional insights regarding the underlying mechanism are necessary. By performing transcriptome analysis coupled with global polar metabolite screening, we show that LXR agonists, LXR623 and GW3965, enhance synergistically the anti‐proliferative effect of BH3 mimetics in solid tumor malignancies, which is predominantly mediated by cell death with features of apoptosis and is rescued by exogenous cholesterol. Extracellular flux analysis and carbon tracing experiments (U‐13C‐glucose and U‐13C‐glutamine) reveal that within 5 h, activation of LXRβ results in reprogramming of tumor cell metabolism, leading to suppression of mitochondrial respiration, a phenomenon not observed in normal human astrocytes. LXR activation elicits a suppression of respiratory complexes at the protein level by reducing their stability. In turn, energy starvation drives an integrated stress response (ISR) that up‐regulates pro‐apoptotic Noxa in an ATF4‐dependent manner. Cholesterol and nucleotides rescue from the ISR elicited by LXR agonists and from cell death induced by LXR agonists and BH3 mimetics. In conventional and patient‐derived xenograft models of colon carcinoma, melanoma, and glioblastoma, the combination treatment of ABT263 and LXR agonists reduces tumor sizes significantly stronger than single treatments. Therefore, the combination treatment of LXR agonists and BH3 mimetics might be a viable efficacious treatment approach for solid malignancies.
- Published
- 2019
- Full Text
- View/download PDF
9. Abnormalities of lipid metabolism in neuronal models of CoQ10 deficiency (S49.004)
- Author
-
Alba Pesini, Giacomo Monzio-Compagnoni, Eliana Barriocanal-Casado, Agustin Hidalgo-Gutierrez, Giulio Kleiner, Giussepe A. Yanez, Mohamed Bakkali, Edoardo Monfrini, Yashpal S. Chhonker, Saba Tadesse, Delfina Larrea, Daryl J. Murry, Caterina Mariotti, Barbara Castellotti, Luis Lopez, Alesio Di Fonzo, Estela Area-Gomez, and Catarina M. Quinzii
- Published
- 2023
10. Supplementary Data from MET Inhibition Elicits PGC1α-Dependent Metabolic Reprogramming in Glioblastoma
- Author
-
Markus D. Siegelin, Peter Canoll, Jeffrey N. Bruce, Georg Karpel-Massler, Catarina M. Quinzii, Mike-Andrew Westhoff, Elena Bianchetti, Giulio Kleiner, Maria J. Sanchez-Quintero, Consuelo Torrini, Chang Shu, Junfei Zhao, Aayushi Mahajan, Nelson Humala, Angeliki Mela, Enyuan Shang, Trang T.T. Nguyen, and Yiru Zhang
- Abstract
Figure S1. Crizotinib resistant cells reveal a reprogrammed tumor metabolism. Figure S2. Crizotinib modulates fatty acid metabolism and mitochondrial parameters. Figure S3. Tracing analysis reveals distinct modulation of metabolism by MET inhibition. Figure S4. Acute and chronic MET inhibition drives oxygen consumption rate and ECAR. Figure S5. Specific silencing of MET enhances oxidative metabolism in patient-derived xenograft cells. Figure S6. Inhibition of complex I and complex V of the electron transport chain and MET synergistically reduce viability of glioblastoma cells. Figure S7. Gamitrinib and etomoxir synergize with crizotinib to reduce cellular viability in glioblastoma model systems. Figure S8. Combined Met inhibition and Gamitrinib treatment elicits cleavage of caspases and modulate the expression of Bcl2 family members. Figure S9. Crizotinib and gamitrinib elicit cell death with features of apoptosis. Figure S10. Gamitrinib and crizotinib is superior over kinase inhibitor combination treatments to elicit apoptosis. Figure S11. The combination treatment of etomoxir and crizotinib elicits a reduction in tumor proliferation without induction of organ toxicity. Supplementary Table 1. Primer sequences for real time PCR and chromatin immunoprecipitation qPCR
- Published
- 2023
11. Data from Combined HDAC and Bromodomain Protein Inhibition Reprograms Tumor Cell Metabolism and Elicits Synthetic Lethality in Glioblastoma
- Author
-
Markus D. Siegelin, Peter Canoll, Georg Karpel-Massler, Mike-Andrew Westhoff, Catarina M. Quinzii, Maria J. Sanchez-Quintero, Giulio Kleiner, Elena Bianchetti, Chang Shu, Junfei Zhao, Sheng-Fu L. Lo, Wataru Ishida, Chiaki Tsuge Ishida, and Yiru Zhang
- Abstract
Purpose: Glioblastoma remains a challenge in oncology, in part due to tumor heterogeneity.Experimental Design: Patient-derived xenograft and stem-like glioblastoma cells were used as the primary model systems.Results: Based on a transcriptome and subsequent gene set enrichment analysis (GSEA), we show by using clinically validated compounds that the combination of histone deacetylase (HDAC) inhibition and bromodomain protein (BRD) inhibition results in pronounced synergistic reduction in cellular viability in patient-derived xenograft and stem-like glioblastoma cells. Transcriptome-based GSEA analysis suggests that metabolic reprogramming is involved with synergistic reduction of oxidative and glycolytic pathways in the combination treatment. Extracellular flux analysis confirms that combined HDAC inhibition and BRD inhibition blunts oxidative and glycolytic metabolism of cancer cells, leading to a depletion of intracellular ATP production and total ATP levels. In turn, energy deprivation drives an integrated stress response, originating from the endoplasmic reticulum. This results in an increase in proapoptotic Noxa. Aside from Noxa, we encounter a compensatory increase of antiapoptotic Mcl-1 protein. Pharmacologic, utilizing the FDA-approved drug sorafenib, and genetic inhibition of Mcl-1 enhanced the effects of the combination therapy. Finally, we show in orthotopic patient-derived xenografts of GBM, that the combination treatment reduces tumor growth, and that triple therapy involving the clinically validated compounds panobinostat, OTX015, and sorafenib further enhances these effects, culminating in a significant regression of tumors in vivo.Conclusions: Overall, these results warrant clinical testing of this novel, efficacious combination therapy. Clin Cancer Res; 24(16); 3941–54. ©2018 AACR.
- Published
- 2023
12. Supplementary Data from Combined HDAC and Bromodomain Protein Inhibition Reprograms Tumor Cell Metabolism and Elicits Synthetic Lethality in Glioblastoma
- Author
-
Markus D. Siegelin, Peter Canoll, Georg Karpel-Massler, Mike-Andrew Westhoff, Catarina M. Quinzii, Maria J. Sanchez-Quintero, Giulio Kleiner, Elena Bianchetti, Chang Shu, Junfei Zhao, Sheng-Fu L. Lo, Wataru Ishida, Chiaki Tsuge Ishida, and Yiru Zhang
- Abstract
Supplementary Data
- Published
- 2023
13. Data from Metabolic Reprogramming by Dual AKT/ERK Inhibition through Imipridones Elicits Unique Vulnerabilities in Glioblastoma
- Author
-
Markus D. Siegelin, Joshua E. Allen, Varun V. Prabhu, Georg Karpel-Massler, Mike-Andrew Westhoff, Catarina M. Quinzii, Maria J. Sanchez-Quintero, Giulio Kleiner, Trang T.T. Nguyen, Chang Shu, Elena Bianchetti, Yiru Zhang, and Chiaki T. Ishida
- Abstract
Purpose: The goal of this study is to enhance the efficacy of imipridones, a novel class of AKT/ERK inhibitors that displayed limited therapeutic efficacy against glioblastoma (GBM).Experimental Design: Gene set enrichment, LC/MS, and extracellular flux analyses were used to determine the mechanism of action of novel imipridone compounds, ONC206 and ONC212. Orthotopic patient-derived xenografts were utilized to evaluate therapeutic potency.Results: Imipridones reduce the proliferation of patient-derived xenograft and stem-like glioblastoma cell cultures in vitro and in multiple xenograft models in vivo. ONC212 displayed the highest potency. High levels of c-myc predict susceptibility to growth inhibition and apoptosis induction by imipridones and increased host survival in orthotopic patient-derived xenografts. As early as 1 hour, imipridones elicit on-target inhibition, followed by dephosphorylation of GSK3β at serine 9. GSK3β promotes phosphorylation of c-myc at threonine 58 and enhances its proteasomal degradation. Moreover, inhibition of c-myc by BRD4 antagonists sensitizes for imipridone-induced apoptosis in stem-like GBM cells in vitro and in vivo. Imipridones affect energy metabolism by suppressing both glycolysis and oxidative phosphorylation, which is accompanied by a compensatory activation of the serine-one carbon-glycine (SOG) pathway, involving the transcription factor ATF4. Interference with the SOG pathway through novel inhibitors of PHGDH results in synergistic cell death induction in vitro and in vivo.Conclusions: These results suggest that c-myc expression predicts therapeutic responses to imipridones and that imipridones lead to suppression of tumor cell energy metabolism, eliciting unique metabolic vulnerabilities that can be exploited for clinical relevant drug combination therapies. Clin Cancer Res; 24(21); 5392–406. ©2018 AACR.
- Published
- 2023
14. Data from MET Inhibition Elicits PGC1α-Dependent Metabolic Reprogramming in Glioblastoma
- Author
-
Markus D. Siegelin, Peter Canoll, Jeffrey N. Bruce, Georg Karpel-Massler, Catarina M. Quinzii, Mike-Andrew Westhoff, Elena Bianchetti, Giulio Kleiner, Maria J. Sanchez-Quintero, Consuelo Torrini, Chang Shu, Junfei Zhao, Aayushi Mahajan, Nelson Humala, Angeliki Mela, Enyuan Shang, Trang T.T. Nguyen, and Yiru Zhang
- Abstract
The receptor kinase c-MET has emerged as a target for glioblastoma therapy. However, treatment resistance emerges inevitably. Here, we performed global metabolite screening with metabolite set enrichment coupled with transcriptome and gene set enrichment analysis and proteomic screening, and identified substantial reprogramming of tumor metabolism involving oxidative phosphorylation and fatty acid oxidation (FAO) with substantial accumulation of acyl-carnitines accompanied by an increase of PGC1α in response to genetic (shRNA and CRISPR/Cas9) and pharmacologic (crizotinib) inhibition of c-MET. Extracellular flux and carbon tracing analyses (U-13C-glucose, U-13C-glutamine, and U-13C-palmitic acid) demonstrated enhanced oxidative metabolism, which was driven by FAO and supported by increased anaplerosis of glucose carbons. These findings were observed in concert with increased number and fusion of mitochondria and production of reactive oxygen species. Genetic interference with PGC1α rescued this oxidative phenotype driven by c-MET inhibition. Silencing and chromatin immunoprecipitation experiments demonstrated that cAMP response elements binding protein regulates the expression of PGC1α in the context of c-MET inhibition. Interference with both oxidative phosphorylation (metformin, oligomycin) and β-oxidation of fatty acids (etomoxir) enhanced the antitumor efficacy of c-MET inhibition. Synergistic cell death was observed with c-MET inhibition and gamitrinib treatment. In patient-derived xenograft models, combination treatments of crizotinib and etomoxir, and crizotinib and gamitrinib were significantly more efficacious than single treatments and did not induce toxicity. Collectively, we have unraveled the mechanistic underpinnings of c-MET inhibition and identified novel combination therapies that may enhance its therapeutic efficacy.Significance:c-MET inhibition causes profound metabolic reprogramming that can be targeted by drug combination therapies.
- Published
- 2023
15. Supplementary Methods from Metabolic Reprogramming by Dual AKT/ERK Inhibition through Imipridones Elicits Unique Vulnerabilities in Glioblastoma
- Author
-
Markus D. Siegelin, Joshua E. Allen, Varun V. Prabhu, Georg Karpel-Massler, Mike-Andrew Westhoff, Catarina M. Quinzii, Maria J. Sanchez-Quintero, Giulio Kleiner, Trang T.T. Nguyen, Chang Shu, Elena Bianchetti, Yiru Zhang, and Chiaki T. Ishida
- Abstract
Supplementary Methods
- Published
- 2023
16. Supplementary Figure Legends from Metabolic Reprogramming by Dual AKT/ERK Inhibition through Imipridones Elicits Unique Vulnerabilities in Glioblastoma
- Author
-
Markus D. Siegelin, Joshua E. Allen, Varun V. Prabhu, Georg Karpel-Massler, Mike-Andrew Westhoff, Catarina M. Quinzii, Maria J. Sanchez-Quintero, Giulio Kleiner, Trang T.T. Nguyen, Chang Shu, Elena Bianchetti, Yiru Zhang, and Chiaki T. Ishida
- Abstract
Supplementary Figure Legends
- Published
- 2023
17. Supplementary Figures 1-9 from Metabolic Reprogramming by Dual AKT/ERK Inhibition through Imipridones Elicits Unique Vulnerabilities in Glioblastoma
- Author
-
Markus D. Siegelin, Joshua E. Allen, Varun V. Prabhu, Georg Karpel-Massler, Mike-Andrew Westhoff, Catarina M. Quinzii, Maria J. Sanchez-Quintero, Giulio Kleiner, Trang T.T. Nguyen, Chang Shu, Elena Bianchetti, Yiru Zhang, and Chiaki T. Ishida
- Abstract
Supplementary Figures
- Published
- 2023
18. Coenzyme Q deficiency causes impairment of the sulfide oxidation pathway
- Author
-
Marcello Ziosi, Ivano Di Meo, Giulio Kleiner, Xing‐Huang Gao, Emanuele Barca, Maria J Sanchez‐Quintero, Saba Tadesse, Hongfeng Jiang, Changhong Qiao, Richard J Rodenburg, Emmanuel Scalais, Markus Schuelke, Belinda Willard, Maria Hatzoglou, Valeria Tiranti, and Catarina M Quinzii
- Subjects
coenzyme Q ,CoQ10 ,Pdss2 ,SQR ,sulfides ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Coenzyme Q (CoQ) is an electron acceptor for sulfide‐quinone reductase (SQR), the first enzyme of the hydrogen sulfide oxidation pathway. Here, we show that lack of CoQ in human skin fibroblasts causes impairment of hydrogen sulfide oxidation, proportional to the residual levels of CoQ. Biochemical and molecular abnormalities are rescued by CoQ supplementation in vitro and recapitulated by pharmacological inhibition of CoQ biosynthesis in skin fibroblasts and ADCK3 depletion in HeLa cells. Kidneys of Pdss2kd/kd mice, which only have ~15% residual CoQ concentrations and are clinically affected, showed (i) reduced protein levels of SQR and downstream enzymes, (ii) accumulation of hydrogen sulfides, and (iii) glutathione depletion. These abnormalities were not present in brain, which maintains ~30% residual CoQ and is clinically unaffected. In Pdss2kd/kd mice, we also observed low levels of plasma and urine thiosulfate and increased blood C4‐C6 acylcarnitines. We propose that impairment of the sulfide oxidation pathway induced by decreased levels of CoQ causes accumulation of sulfides and consequent inhibition of short‐chain acyl‐CoA dehydrogenase and glutathione depletion, which contributes to increased oxidative stress and kidney failure.
- Published
- 2016
- Full Text
- View/download PDF
19. The clinical heterogeneity of coenzyme Q10 deficiency results from genotypic differences in the Coq9 gene
- Author
-
Marta Luna‐Sánchez, Elena Díaz‐Casado, Emanuele Barca, Miguel Ángel Tejada, Ángeles Montilla‐García, Enrique Javier Cobos, Germaine Escames, Dario Acuña‐Castroviejo, Catarina M Quinzii, and Luis Carlos López
- Subjects
CoQ multiprotein complex ,Coq9 ,mitochondrial myopathy ,mouse model ,nonsense‐mediated mRNA decay ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Primary coenzyme Q10 (CoQ10) deficiency is due to mutations in genes involved in CoQ biosynthesis. The disease has been associated with five major phenotypes, but a genotype–phenotype correlation is unclear. Here, we compare two mouse models with a genetic modification in Coq9 gene (Coq9Q95X and Coq9R239X), and their responses to 2,4‐dihydroxybenzoic acid (2,4‐diHB). Coq9R239X mice manifest severe widespread CoQ deficiency associated with fatal encephalomyopathy and respond to 2,4‐diHB increasing CoQ levels. In contrast, Coq9Q95X mice exhibit mild CoQ deficiency manifesting with reduction in CI+III activity and mitochondrial respiration in skeletal muscle, and late‐onset mild mitochondrial myopathy, which does not respond to 2,4‐diHB. We show that these differences are due to the levels of COQ biosynthetic proteins, suggesting that the presence of a truncated version of COQ9 protein in Coq9R239X mice destabilizes the CoQ multiprotein complex. Our study points out the importance of the multiprotein complex for CoQ biosynthesis in mammals, which may provide new insights to understand the genotype–phenotype heterogeneity associated with human CoQ deficiency and may have a potential impact on the treatment of this mitochondrial disorder.
- Published
- 2015
- Full Text
- View/download PDF
20. Deoxypyrimidine monophosphate bypass therapy for thymidine kinase 2 deficiency
- Author
-
Caterina Garone, Beatriz Garcia‐Diaz, Valentina Emmanuele, Luis C Lopez, Saba Tadesse, Hasan O Akman, Kurenai Tanji, Catarina M Quinzii, and Michio Hirano
- Subjects
deoxycytidine monophosphate ,deoxythymidine monophosphate ,encephalomyopathy ,therapy ,thymidine kinase ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Autosomal recessive mutations in the thymidine kinase 2 gene (TK2) cause mitochondrial DNA depletion, multiple deletions, or both due to loss of TK2 enzyme activity and ensuing unbalanced deoxynucleotide triphosphate (dNTP) pools. To bypass Tk2 deficiency, we administered deoxycytidine and deoxythymidine monophosphates (dCMP+dTMP) to the Tk2 H126N (Tk2−/−) knock‐in mouse model from postnatal day 4, when mutant mice are phenotypically normal, but biochemically affected. Assessment of 13‐day‐old Tk2−/− mice treated with dCMP+dTMP 200 mg/kg/day each (Tk2−/−200dCMP/dTMP) demonstrated that in mutant animals, the compounds raise dTTP concentrations, increase levels of mtDNA, ameliorate defects of mitochondrial respiratory chain enzymes, and significantly prolong their lifespan (34 days with treatment versus 13 days untreated). A second trial of dCMP+dTMP each at 400 mg/kg/day showed even greater phenotypic and biochemical improvements. In conclusion, dCMP/dTMP supplementation is the first effective pharmacologic treatment for Tk2 deficiency.
- Published
- 2014
- Full Text
- View/download PDF
21. Inefficient thermogenic mitochondrial respiration due to futile proton leak in a mouse model of fragile X syndrome
- Author
-
Linlin Sun, Catarina M. Quinzii, Matthew Tracey, Elizabeth A. Jonas, Jose F. Perez-Zoghbi, Pawel Licznerski, Giulio Kleiner, Aili Wang, Richard J. Levy, Mu Yang, Lifei Wang, Guang Yang, and Keren K. Griffiths
- Subjects
Male ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Neurogenesis ,Cell Respiration ,Synaptogenesis ,Oxidative phosphorylation ,Mitochondrion ,Biology ,Biochemistry ,Article ,Fragile X Mental Retardation Protein ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Intellectual Disability ,Genetics ,medicine ,Animals ,Autistic Disorder ,Molecular Biology ,Mice, Knockout ,Thermogenesis ,medicine.disease ,FMR1 ,Mitochondria ,Cell biology ,Fragile X syndrome ,Disease Models, Animal ,030104 developmental biology ,Fragile X Syndrome ,Coenzyme Q – cytochrome c reductase ,Knockout mouse ,Forebrain ,Female ,Protons ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Fragile X syndrome (FXS) is the leading known inherited intellectual disability and the most common genetic cause of autism. The full mutation results in transcriptional silencing of the Fmr1 gene and loss of fragile X mental retardation protein (FMRP) expression. Defects in neuroenergetic capacity are known to cause a variety of neurodevelopmental disorders. Thus, we explored the integrity of forebrain mitochondria in Fmr1 knockout mice during the peak of synaptogenesis. We found inefficient thermogenic respiration due to futile proton leak in Fmr1 KO mitochondria caused by coenzyme Q (CoQ) deficiency and an open cyclosporine-sensitive channel. Repletion of mitochondrial CoQ within the Fmr1 KO forebrain closed the channel, blocked the pathological proton leak, restored rates of protein synthesis during synaptogenesis, and normalized the key phenotypic features later in life. The findings demonstrate that FMRP deficiency results in inefficient oxidative phosphorylation during the neurodevelopment and suggest that dysfunctional mitochondria may contribute to the FXS phenotype.
- Published
- 2020
22. Redefining infantile-onset multisystem phenotypes of coenzyme Q10-deficiency in the next-generation sequencing era
- Author
-
Andres Berardo and Catarina M. Quinzii
- Subjects
Whole genome sequencing ,Coenzyme Q10 ,business.industry ,nephrotic syndrome ,Encephalopathy ,coenzyme Q biosynthesis ,medicine.disease ,Bioinformatics ,encephalopathy ,Phenotype ,coenzyme Q10 deficiency ,Article ,cardiopathy ,chemistry.chemical_compound ,chemistry ,medicine ,Coenzyme Q10 deficiency ,business ,Gene ,Nephrotic syndrome ,Exome sequencing - Abstract
Primary coenzyme Q10 (CoQ10) deficiency encompasses a subset of mitochondrial diseases caused by mutations affecting proteins involved in the CoQ10 biosynthetic pathway. One of the most frequent clinical syndromes associated with primary CoQ10 deficiency is the severe infantile multisystemic form, which, until recently, was underdiagnosed. In the last few years, the availability of genetic screening through whole exome sequencing and whole genome sequencing has enabled molecular diagnosis in a growing number of patients with this syndrome and has revealed new disease phenotypes and molecular defects in CoQ10 biosynthetic pathway genes. Early genetic screening can rapidly and non-invasively diagnose primary CoQ10 deficiencies. Early diagnosis is particularly important in cases of CoQ10 deficient steroid-resistant nephrotic syndrome, which frequently improves with treatment. In contrast, the infantile multisystemic forms of CoQ10 deficiency, particularly when manifesting with encephalopathy, present therapeutic challenges, due to poor responses to CoQ10 supplementation. Administration of CoQ10 biosynthetic intermediate compounds is a promising alternative to CoQ10; however, further pre-clinical studies are needed to establish their safety and efficacy, as well as to elucidate the mechanism of actions of the intermediates. Here, we review the molecular defects causes of the multisystemic infantile phenotype of primary CoQ10 deficiency, genotype-phenotype correlations, and recent therapeutic advances.
- Published
- 2020
23. Aurora kinase A inhibition reverses the Warburg effect and elicits unique metabolic vulnerabilities in glioblastoma
- Author
-
Sungsoo Kim, Trang T. T. Nguyen, Mike-Andrew Westhoff, Catarina M. Quinzii, Hee Won Yang, Peter Canoll, Chang Shu, Hasan O. Akman, Nelson Humala, Jeffrey N. Bruce, Georg Karpel-Massler, Angeliki Mela, Enyuan Shang, Guoan Zhang, Markus D. Siegelin, and Aayushi Mahajan
- Subjects
Proteomics ,Science ,General Physics and Astronomy ,Article ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,Cell Line, Tumor ,Coactivator ,Warburg Effect, Oncologic ,Humans ,PPAR alpha ,Glycolysis ,Receptor ,Aurora Kinase A ,Cell Proliferation ,Multidisciplinary ,Chemistry ,Fatty Acids ,General Chemistry ,Cancer metabolism ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Warburg effect ,nervous system diseases ,Chromatin ,CNS cancer ,Cancer research ,Peroxisome proliferator-activated receptor alpha ,Glioblastoma ,Signal Transduction - Abstract
Aurora kinase A (AURKA) has emerged as a drug target for glioblastoma (GBM). However, resistance to therapy remains a critical issue. By integration of transcriptome, chromatin immunoprecipitation sequencing (CHIP-seq), Assay for Transposase-Accessible Chromatin sequencing (ATAC-seq), proteomic and metabolite screening followed by carbon tracing and extracellular flux analyses we show that genetic and pharmacological AURKA inhibition elicits metabolic reprogramming mediated by inhibition of MYC targets and concomitant activation of Peroxisome Proliferator Activated Receptor Alpha (PPARA) signaling. While glycolysis is suppressed by AURKA inhibition, we note an increase in the oxygen consumption rate fueled by enhanced fatty acid oxidation (FAO), which was accompanied by an increase of Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α). Combining AURKA inhibitors with inhibitors of FAO extends overall survival in orthotopic GBM PDX models. Taken together, these data suggest that simultaneous targeting of oxidative metabolism and AURKAi might be a potential novel therapy against recalcitrant malignancies., Glioblastoma patients are treated with Aurora kinase A (AURKA) inhibitors but resistance can occur. Here, the authors show that AURKA inhibition induces metabolic reprogramming, which leads to increased mitochondrial activity and inhibition of oxidative metabolism sensitizes glioblastoma cells to AURKA inhibition.
- Published
- 2021
24. Dystonia-Ataxia with early handwriting deterioration in COQ8A mutation carriers: A case series and literature review
- Author
-
Gessica Vasco, Filippo M. Santorelli, Enrico Bertini, Maria Lieto, Tommaso Schirinzi, Jennifer Friedman, Vincenzo Leuzzi, Michio Hirano, Serena Galosi, Alessandro Filla, Richard H. Haas, Ginevra Zanni, Rosalba Carrozzo, Catarina M. Quinzii, Daniele Galatolo, Michela Di Nottia, and Emanuele Barca
- Subjects
Adult ,Male ,0301 basic medicine ,Handwriting ,Heterozygote ,congenital, hereditary, and neonatal diseases and abnormalities ,Pediatrics ,medicine.medical_specialty ,Mitochondrial Diseases ,Ataxia ,Dystonia-ataxia syndrome ,Ubiquinone ,COQ8A ,Disease ,Mitochondrial Proteins ,Young Adult ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Handwriting deterioration ,Humans ,Medicine ,Child ,Dystonia ,CoQ ,10 ,deficiency ,Female ,Middle Aged ,Disease Progression ,Dystonic Disorders ,Muscle Weakness ,Cerebellar ataxia ,business.industry ,Focal dystonia ,medicine.disease ,Natural history ,030104 developmental biology ,Neurology ,Speech disorder ,Neurology (clinical) ,Geriatrics and Gerontology ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Cerebellar ataxia is a hallmark of coenzyme Q10 (CoQ10) deficiency associated with COQ8A mutations. We present four patients, one with novel COQ8A pathogenic variants all with early, prominent handwriting impairment, dystonia and only mild ataxia. To better define the phenotypic spectrum and course of COQ8A disease, we review the clinical presentation and evolution in 47 reported cases. Individuals with COQ8A mutation display great clinical variability and unpredictable responses to CoQ10 supplementation. Onset is typically during infancy or childhood with ataxic features associated with developmental delay or regression. When disease onset is later in life, first symptoms can include: incoordination, epilepsy, tremor, and deterioration of writing. The natural history is characterized by a progression to a multisystem brain disease dominated by ataxia, with disease severity inversely correlated with age at onset. Six previously reported cases share with ours, a clinical phenotype characterized by slowly progressive or static writing difficulties, focal dystonia, and speech disorder, with only minimal ataxia. The combination of writing difficulty, dystonia and ataxia is a distinctive constellation that is reminiscent of a previously described clinical entity called Dystonia Ataxia Syndrome (DYTCA) and is an important clinical indicator of COQ8A mutations, even when ataxia is mild or absent.
- Published
- 2019
25. Abnormalities of hydrogen sulfide and glutathione pathways in mitochondrial dysfunction
- Author
-
Luis C. López and Catarina M. Quinzii
- Subjects
0301 basic medicine ,Mitochondrial disease ,Mitochondrion ,Bioinformatics ,medicine.disease_cause ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Biosynthesis ,medicine ,lcsh:Science (General) ,ComputingMethodologies_COMPUTERGRAPHICS ,lcsh:R5-920 ,Multidisciplinary ,Mechanism (biology) ,business.industry ,Reactive oxygen species (ROS) ,ROS ,Coenzyme Q ,Glutathione ,Metabolism ,medicine.disease ,equipment and supplies ,3. Good health ,Mitochondria ,030104 developmental biology ,chemistry ,Oxidative stress ,030220 oncology & carcinogenesis ,Coenzyme Q – cytochrome c reductase ,lcsh:Medicine (General) ,business ,lcsh:Q1-390 - Abstract
This work was supported by NIH P01 HD080642 (CMQ), and Ministerio de Ciencia e inn (LCL). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript., Background Mitochondrial disorders are genetic diseases for which therapy remains woefully inadequate. Therapy of these disorders is particularly challenging partially due to the heterogeneity and tissue-specificity of pathomechanisms involved in these disorders. Abnormalities in hydrogen sulfide (H2S) metabolism are emerging as novel mechanism in mitochondrial dysfunction. However, further studies are necessary to understand the effects, protective or detrimental, of these abnormalities, and their relevance, in mitochondrial diseases. Aim of Review: To review the recent evidences of derangement of the metabolism of H2S, at biosynthesis or oxidation levels, in mitochondrial dysfunction, focusing specifically on the alterations of H2S oxidation caused by primary Coenzyme Q (CoQ) deficiency. Key Scientific Concepts of Review: Mitochondria play a key role in the regulation of H2S and GSH metabolism pathways. However, further studies are needed to understand the consequences of abnormalities of H2S and GSH synthesis on the oxidation pathway, and vice versa; and on the levels of H2S and GSH, their tissue-specific detrimental effects, and their role the role in mitochondrial diseases. Beside the known H2S pathways, additional, tissue-specific, enzymatic systems, involved in H2S production and elimination, might exist., United States Department of Health & Human Services National Institutes of Health (NIH) - USA P01 HD080642, Ministerio de Ciencia e inn (LCL)
- Published
- 2021
- Full Text
- View/download PDF
26. Therapies Approaches in Mitochondrial Diseases
- Author
-
Catarina M. Quinzii, Valentina Emmanuele, and Michio Hirano
- Subjects
Clinical trial ,Mitochondrial DNA ,Mitochondrial biogenesis ,Mitochondrial replacement therapy ,business.industry ,Mitophagy ,medicine ,Cell replacement ,Hypoxia (medical) ,medicine.symptom ,Bioinformatics ,business ,Heteroplasmy - Abstract
Therapies for mitochondrial diseases has been largely limited to supportive and symptomatic therapies; however, in the last decade, advances in understanding the causes and pathomechanisms of these diverse disorders have enabled development of novel treatment strategies. Here, we highlight current use of dietary supplements and exercise therapy as well as emerging treatments in preclinical and clinical trial stages of development. Broad-spectrum therapies that may be applied multiple diseases include: activation of mitochondrial biogenesis, regulation of mitophagy and mitochondrial dynamics, bypass of mitochondrial biochemical defects, mitochondrial replacement therapy, and hypoxia. Tailored disease-specific therapies in development include: scavenging of toxic compounds, deoxynucleoside therapy, cell replacement therapies, viral-mediated gene-delivery, shifting heteroplasmy of mitochondrial DNA pathogenic variants, and stabilization of mitochondrial transfer RNAs.
- Published
- 2021
27. Regulatory environment for novel therapeutic development in mitochondrial diseases
- Author
-
Valentina Emmanuele, Emanuele Barca, Michio Hirano, Camilla V Simpson, Xiomara Q Rosales, Andres Berardo, John L.P. Thompson, Kristin Engelstad, and Catarina M. Quinzii
- Subjects
Flexibility (engineering) ,medicine.medical_specialty ,Mitochondrial Diseases ,Orphan Drug Production ,business.industry ,Surrogate endpoint ,United States Food and Drug Administration ,Legislation ,Patient advocacy ,United States ,Article ,Orphan drug ,Clinical trial ,Rare Diseases ,Genetics ,Medicine ,media_common.cataloged_instance ,Humans ,European union ,business ,Intensive care medicine ,Drug Approval ,Genetics (clinical) ,Pharmaceutical industry ,media_common - Abstract
At present, there is just one approved therapy for patients with mitochondrial diseases in Europe, another in Japan, and none in the United States. These facts reveal an important and significant unmet need for approved therapies for these debilitating and often fatal disorders. To fill this need, it is critical for clinicians and drug developers to work closely with regulatory agencies. In the United States, mitochondrial disease patients and clinicians, the United Mitochondrial Disease Foundation, and pharmaceutical industry members have engaged with the Food and Drug Administration to educate each other about these complex and heterogeneous diseases and about regulatory requirements to obtain approvals for novel therapies. Clinical development of therapies for rare diseases has been facilitated by the 1983 US Orphan Drug Act (ODA) and similar legislation in Japan and the European Union. Further legislation and regulatory guidance have expanded and refined regulatory flexibility. While regulatory and financial incentives of the ODA have augmented involvement of pharmaceutical companies, clinicians, with patient advocacy groups and industry, need to conduct natural history studies, develop clinical outcome measures, and identify potential supportive surrogate endpoints predictive of clinical benefit, which together are critical foundations for clinical trials. Thus, the regulatory environment for novel therapeutic development is conducive and offers flexibility for mitochondrial diseases. Nevertheless, flexibility does not mean lower standards, as well-controlled rigorous clinical trials of high quality are still required to establish the efficacy of potential therapies and to obtain regulatory agency approvals for their commercial use. This process is illustrated through the authors' ongoing efforts to develop therapy for thymidine kinase 2 deficiency.
- Published
- 2020
28. TAMI-33. AURKA INHIBITION REPROGRAMS METABOLISM AND IS SYNTHETICALLY LETHAL WITH FATTY ACID OXIDATION INHIBITION IN GLIOBLASTOMA
- Author
-
Markus D. Siegelin, Aayushi Mahajan, Catarina M. Quinzii, Trang T. T. Nguyen, Georg Karpel-Massler, Enyuan Shang, Nelson Humala, Angeliki Mela, Mike-Andrew Westhoff, Peter Canoll, Chang Shu, and Jeffrey N. Bruce
- Subjects
Cancer Research ,Oncology ,Biochemistry ,Chemistry ,medicine ,Tumor Microenvironment/Angiogenesis/Metabolism/Invasion ,Neurology (clinical) ,Metabolism ,medicine.disease ,Beta oxidation ,Glioblastoma - Abstract
Aurora kinase A (AURKA) has emerged as a viable drug target for glioblastoma (GBM), the most common malignant primary brain tumor in adults with a life expectancy of 12-15 months. However, resistance to therapy remains a critical issue, which partially may be driven by reprogramming of metabolism. By integration of transcriptome, chromatin immunoprecipitation with sequencing (CHIP-seq.), assay for transposase-accessible chromatin with sequencing (ATAC-seq.), proteomic and metabolite screening followed by carbon tracing (U-13C-Glucose, U-13C-Glutamine and U-13C-Palmitic acid) and extracellular flux analysis we provided evidence that genetic (shRNA and CRISPR/Cas9) and pharmacological (Alisertib) AURKA inhibition elicited substantial metabolic reprogramming mediated in part by inhibition of MYC targets and concomitant activation of PPARA (e.g. PGC1A) signaling. While glycolysis was suppressed by AURKA inhibition, we noted a compensatory increase in oxygen consumption rate (OCR) fueled by fatty acid oxidation (FAO). Whereas interference with AURKA elicited a suppression of c-Myc, we detected an upregulation of PGC1A, a master regulator of oxidative metabolism, upon AURKA inhibition. Silencing of PGC1A reversed the increase in OCR and sensitized GBM cells to AURKA inhibition mediated reduction in cellular viability. CHIP experiments confirmed binding of c-Myc to the promoter region of PGC1A, which is abrogated by AURKA inhibition and in turn unleashed PGC1A expression. ATAC-seq. confirmed higher accessibility of the MYC binding region within the PGC1A promoter. Forced expression of c-Myc blocked AURKA inhibition mediated increase of PGC1A, suggesting that c-Myc acted as a repressor. To interfere with this oxidative metabolic reprogramming, we combined AURKA inhibitors with blockers of FAO (etomoxir), which elicited substantial synergistic growth inhibition and extension of overall survival in orthotopic patient derived xenografts of GBM in mice without induction of toxicity in normal tissue. Taken together, these data support that simultaneous targeting of oxidative metabolism and AURKA inhibition might be a potential novel therapy against GBM.
- Published
- 2020
29. Inhibition of HDAC1/2 Along with TRAP1 Causes Synthetic Lethality in Glioblastoma Model Systems
- Author
-
Markus D. Siegelin, Enyuan Shang, Chang Shu, Yiru Zhang, Trang T. T. Nguyen, Catarina M. Quinzii, Mike-Andrew Westhoff, and Georg Karpel-Massler
- Subjects
Programmed cell death ,Chromatin Immunoprecipitation ,Cell Survival ,Blotting, Western ,Histone Deacetylase 2 ,Antineoplastic Agents ,Histone Deacetylase 1 ,Synthetic lethality ,Real-Time Polymerase Chain Reaction ,Article ,Romidepsin ,Electron Transport ,chemistry.chemical_compound ,Mice ,HDAC inhibitors ,Bcl-2 family ,Panobinostat ,gamitrinib ,Cell Line, Tumor ,In Situ Nick-End Labeling ,Medicine ,Animals ,Humans ,HSP90 Heat-Shock Proteins ,RNA, Small Interfering ,lcsh:QH301-705.5 ,Caspase ,biology ,business.industry ,Histone deacetylase 2 ,glioblastoma ,electron transport chain ,General Medicine ,Flow Cytometry ,Xenograft Model Antitumor Assays ,Histone Deacetylase Inhibitors ,chemistry ,lcsh:Biology (General) ,Proto-Oncogene Proteins c-bcl-2 ,Apoptosis ,Cancer research ,biology.protein ,tumor metabolism ,business ,medicine.drug - Abstract
The heterogeneity of glioblastomas, the most common primary malignant brain tumor, remains a significant challenge for the treatment of these devastating tumors. Therefore, novel combination treatments are warranted. Here, we showed that the combined inhibition of TRAP1 by gamitrinib and histone deacetylases (HDAC1/HDAC2) through romidepsin or panobinostat caused synergistic growth reduction of established and patient-derived xenograft (PDX) glioblastoma cells. This was accompanied by enhanced cell death with features of apoptosis and activation of caspases. The combination treatment modulated the levels of pro- and anti-apoptotic Bcl-2 family members, including BIM and Noxa, Mcl-1, Bcl-2 and Bcl-xL. Silencing of Noxa, BAK and BAX attenuated the effects of the combination treatment. At the metabolic level, the combination treatment led to an enhanced reduction of oxygen consumption rate and elicited an unfolded stress response. Finally, we tested whether the combination treatment of gamitrinib and panobinostat exerted therapeutic efficacy in PDX models of glioblastoma (GBM) in mice. While single treatments led to mild to moderate reduction in tumor growth, the combination treatment suppressed tumor growth significantly stronger than single treatments without induction of toxicity. Taken together, we have provided evidence that simultaneous targeting of TRAP1 and HDAC1/2 is efficacious to reduce tumor growth in model systems of glioblastoma.
- Published
- 2020
30. Targeting a Braf/Mapk pathway rescues podocyte lipid peroxidation in CoQ-deficiency kidney disease
- Author
-
Catarina M. Quinzii, Giulio Kleiner, Clary B. Clish, Katherine A. Vernon, Keith Keller, Anna Greka, Choah Kim, May Theng Ting, Astrid Weins, Andrew J.B. Watts, Nicolas Wieder, Eriene-Heidi Sidhom, Maria Kost-Alimova, Matthew Racette, Elizabeth J. Grinkevich, Jillian L. Shaw, Estefanía Reyes-Bricio, Jamie L. Marshall, Fei Chen, and Julian Avila-Pacheco
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Nephrology ,Proto-Oncogene Proteins B-raf ,medicine.medical_specialty ,Mitochondrial Diseases ,MAP Kinase Signaling System ,Ubiquinone ,Mitochondrion ,GPX4 ,Podocyte ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Drug Delivery Systems ,PDSS2 ,Internal medicine ,medicine ,Animals ,Humans ,RNA-Seq ,Kidney ,Alkyl and Aryl Transferases ,Muscle Weakness ,Chemistry ,Podocytes ,food and beverages ,General Medicine ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,Indenes ,030220 oncology & carcinogenesis ,Cancer research ,Pyrazoles ,Ataxia ,Kidney Diseases ,Lipid Peroxidation ,Kidney disease ,Research Article - Abstract
Mutations affecting mitochondrial coenzyme Q (CoQ) biosynthesis lead to kidney failure due to selective loss of podocytes, essential cells of the kidney filter. Curiously, neighboring tubular epithelial cells are spared early in disease despite higher mitochondrial content. We sought to illuminate noncanonical, cell-specific roles for CoQ, independently of the electron transport chain (ETC). Here, we demonstrate that CoQ depletion caused by Pdss2 enzyme deficiency in podocytes results in perturbations in polyunsaturated fatty acid (PUFA) metabolism and the Braf/Mapk pathway rather than ETC dysfunction. Single-nucleus RNA-Seq from kidneys of Pdss2(kd/kd) mice with nephrotic syndrome and global CoQ deficiency identified a podocyte-specific perturbation of the Braf/Mapk pathway. Treatment with GDC-0879, a Braf/Mapk-targeting compound, ameliorated kidney disease in Pdss2(kd/kd) mice. Mechanistic studies in Pdss2-depleted podocytes revealed a previously unknown perturbation in PUFA metabolism that was confirmed in vivo. Gpx4, an enzyme that protects against PUFA-mediated lipid peroxidation, was elevated in disease and restored after GDC-0879 treatment. We demonstrate broader human disease relevance by uncovering patterns of GPX4 and Braf/Mapk pathway gene expression in tissue from patients with kidney diseases. Our studies reveal ETC-independent roles for CoQ in podocytes and point to Braf/Mapk as a candidate pathway for the treatment of kidney diseases.
- Published
- 2020
31. Coenzyme Q10 modulates sulfide metabolism and links the mitochondrial respiratory chain to pathways associated to one carbon metabolism
- Author
-
Sabina Sánchez-Hernández, Luis C. López, Pilar González-García, Catarina M. Quinzii, Francisco Martin, Holger Prokisch, Ussipbek Botagoz Abdihankyzy, Agustín Hidalgo-Gutiérrez, Cristina Mascaraque, Germaine Escames, Marcello Ziosi, Mohammed Bakkali, and Eliana Barriocanal-Casado
- Subjects
AcademicSubjects/SCI01140 ,Mitochondrial Diseases ,Sulfide ,Ubiquinone ,Respiratory chain ,Transsulfuration ,Transsulfuration pathway ,Mitochondrion ,Biology ,Sulfides ,Electron Transport ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genetics ,Animals ,Humans ,Oxidoreductases Acting on Sulfur Group Donors ,Molecular Biology ,Genetics (clinical) ,030304 developmental biology ,Skin ,chemistry.chemical_classification ,0303 health sciences ,Electron Transport Complex I ,Muscle Weakness ,General Medicine ,Metabolism ,Vitamins ,Fibroblasts ,Cystathionine beta synthase ,Glutathione ,Carbon ,3. Good health ,Mitochondria ,Mice, Inbred C57BL ,Mitochondrial respiratory chain ,Biochemistry ,chemistry ,biology.protein ,Ataxia ,General Article ,Transcriptome ,030217 neurology & neurosurgery - Abstract
This work was supported by grants from Ministerio de Ciencia e Innovacion, Spain, and the ERDF (RTI2018-093503-B-100); the Muscular Dystrophy Association (MDA-602322); the University of Granada (grant reference 'UNETE', UCE-PP2017-06) (L.C.L.) and the National Institute of Health (NIH, United States) P01 HD080642-01 (C.M.Q.). A.H.-G. and P.G.-G. are `FPU fellows' from the Ministerio de Universidades, Spain. E.B.-C. was supported by the Junta de Andalucia. U.B.A. was supported by the Erasmus+ Program., Abnormalities of one carbon, glutathione and sulfide metabolisms have recently emerged as novel pathomechanisms in diseases with mitochondrial dysfunction. However, the mechanisms underlying these abnormalities are not clear. Also, we recently showed that sulfide oxidation is impaired in Coenzyme Q10 (CoQ10) deficiency. This finding leads us to hypothesize that the therapeutic effects of CoQ10, frequently administered to patients with primary or secondary mitochondrial dysfunction, might be due to its function as cofactor for sulfide:quinone oxidoreductase (SQOR), the first enzyme in the sulfide oxidation pathway. Here, using biased and unbiased approaches, we show that supraphysiological levels of CoQ10 induces an increase in the expression of SQOR in skin fibroblasts from control subjects and patients with mutations in Complex I subunits genes or CoQ biosynthetic genes. This increase of SQOR induces the downregulation of the cystathionine β-synthase and cystathionine γ-lyase, two enzymes of the transsulfuration pathway, the subsequent downregulation of serine biosynthesis and the adaptation of other sulfide linked pathways, such as folate cycle, nucleotides metabolism and glutathione system. These metabolic changes are independent of the presence of sulfur aminoacids, are confirmed in mouse models, and are recapitulated by overexpression of SQOR, further proving that the metabolic effects of CoQ10 supplementation are mediated by the overexpression of SQOR. Our results contribute to a better understanding of how sulfide metabolism is integrated in one carbon metabolism and may explain some of the benefits of CoQ10 supplementation observed in mitochondrial diseases., Spanish Government, European Union (EU) RTI2018-093503-B-100, Muscular Dystrophy Association MDA-602322, University of Granada UCE-PP2017-06, United States Department of Health & Human Services National Institutes of Health (NIH) - USA P01 HD080642-01, Junta de Andalucia, Erasmus+ Program
- Published
- 2020
32. Metabolic Reprogramming by Dual AKT/ERK Inhibition through Imipridones Elicits Unique Vulnerabilities in Glioblastoma
- Author
-
Mike-Andrew Westhoff, Maria J. Sanchez-Quintero, Chiaki Tsuge Ishida, Joshua E. Allen, Chang Shu, Georg Karpel-Massler, Giulio Kleiner, Catarina M. Quinzii, Yiru Zhang, Varun V. Prabhu, Elena Bianchetti, Trang T. T. Nguyen, and Markus D. Siegelin
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Cell Survival ,Antineoplastic Agents ,Apoptosis ,Oxidative Phosphorylation ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,In vivo ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase B ,Cell Proliferation ,Membrane Potential, Mitochondrial ,Chemistry ,ATF4 ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Mechanism of action ,Cancer research ,Phosphorylation ,medicine.symptom ,Growth inhibition ,Energy Metabolism ,Glioblastoma ,Glycolysis ,Protein Processing, Post-Translational ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Purpose: The goal of this study is to enhance the efficacy of imipridones, a novel class of AKT/ERK inhibitors that displayed limited therapeutic efficacy against glioblastoma (GBM). Experimental Design: Gene set enrichment, LC/MS, and extracellular flux analyses were used to determine the mechanism of action of novel imipridone compounds, ONC206 and ONC212. Orthotopic patient-derived xenografts were utilized to evaluate therapeutic potency. Results: Imipridones reduce the proliferation of patient-derived xenograft and stem-like glioblastoma cell cultures in vitro and in multiple xenograft models in vivo. ONC212 displayed the highest potency. High levels of c-myc predict susceptibility to growth inhibition and apoptosis induction by imipridones and increased host survival in orthotopic patient-derived xenografts. As early as 1 hour, imipridones elicit on-target inhibition, followed by dephosphorylation of GSK3β at serine 9. GSK3β promotes phosphorylation of c-myc at threonine 58 and enhances its proteasomal degradation. Moreover, inhibition of c-myc by BRD4 antagonists sensitizes for imipridone-induced apoptosis in stem-like GBM cells in vitro and in vivo. Imipridones affect energy metabolism by suppressing both glycolysis and oxidative phosphorylation, which is accompanied by a compensatory activation of the serine-one carbon-glycine (SOG) pathway, involving the transcription factor ATF4. Interference with the SOG pathway through novel inhibitors of PHGDH results in synergistic cell death induction in vitro and in vivo. Conclusions: These results suggest that c-myc expression predicts therapeutic responses to imipridones and that imipridones lead to suppression of tumor cell energy metabolism, eliciting unique metabolic vulnerabilities that can be exploited for clinical relevant drug combination therapies. Clin Cancer Res; 24(21); 5392–406. ©2018 AACR.
- Published
- 2018
33. CoQ10 supplementation rescues nephrotic syndrome through normalization of H2S oxidation pathway
- Author
-
Giulio Kleiner, Agustín Hidalgo-Gutiérrez, Luis C. López, Emanuele Barca, Valentina Emmanuele, Saba Tadesse, Yimeng Xu, Marcello Ziosi, Estela Area-Gomez, Changhong Qiao, and Catarina M. Quinzii
- Subjects
0301 basic medicine ,Mitochondrial Diseases ,Nephrotic Syndrome ,Antioxidant ,Ubiquinone ,medicine.medical_treatment ,Mitochondrion ,Kidney ,medicine.disease_cause ,Antioxidants ,Mice ,chemistry.chemical_compound ,Oxidoreductases Acting on Sulfur Group Donors ,Hydrogen Sulfide ,chemistry.chemical_classification ,Muscle Weakness ,Chemistry ,food and beverages ,Mitochondria ,Coenzyme Q(10) ,medicine.anatomical_structure ,Molecular Medicine ,Oxidation-Reduction ,Metabolic Networks and Pathways ,medicine.medical_specialty ,Mice, Transgenic ,Sulfides ,Article ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Humans ,Molecular Biology ,Coenzyme Q10 ,Reactive oxygen species ,Alkyl and Aryl Transferases ,Metabolism ,medicine.disease ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,Ataxia ,CoQ deficiency ,Reactive Oxygen Species ,Oxidative stress ,HeLa Cells ,Kidney disease - Abstract
Nephrotic syndrome (NS), a frequent chronic kidney disease in children and young adults, is the most common phenotype associated with primary coenzyme Q(10) (CoQ(10)) deficiency and is very responsive to CoQ(10) supplementation, although the pathomechanism is not clear. Here, using a mouse model of CoQ deficiency-associated NS, we show that long-term oral CoQ(10) supplementation prevents kidney failure by rescuing defects of sulfides oxidation and ameliorating oxidative stress, despite only incomplete normalization of kidney CoQ levels and lack of rescue of CoQ-dependent respiratory enzymes activities. Liver and kidney lipidomics, and urine metabolomics analyses, did not show CoQ metabolites. To further demonstrate that sulfides metabolism defects cause oxidative stress in CoQ deficiency, we show that silencing of sulfide quinone oxido-reductase (SQOR) in wild-type HeLa cells leads to similar increases of reactive oxygen species (ROS) observed in HeLa cells depleted of the CoQ biosynthesis regulatory protein COQ8A. While CoQ(10) supplementation of COQ8A depleted cells decreases ROS and increases SQOR protein levels, knock-down of SQOR prevents CoQ(10) antioxidant effects. We conclude that kidney failure in CoQ deficiency-associated NS is caused by oxidative stress mediated by impaired sulfides oxidation and propose that CoQ supplementation does not significantly increase the kidney pool of CoQ bound to the respiratory supercomplexes, but rather enhances the free pool of CoQ, which stabilizes SQOR protein levels rescuing oxidative stress.
- Published
- 2018
34. Cardiomyopathy and altered integrin-actin signaling in Fhl1 mutant female mice
- Author
-
Maria J. Sanchez-Quintero, Fusako Sera, Shunichi Homma, Valentina Emmanuele, Catarina M. Quinzii, Shingo Kariya, Martí Juanola-Falgarona, Kurenai Tanji, Akatsuki Kubota, and Michio Hirano
- Subjects
Male ,Proteomics ,Heterozygote ,Integrins ,medicine.medical_specialty ,Mutant ,Mutation, Missense ,Cardiomyopathy ,Muscle Proteins ,Biology ,Mice ,03 medical and health sciences ,Muscular Diseases ,Internal medicine ,Genetics ,medicine ,Animals ,Muscular dystrophy ,Muscle, Skeletal ,Myopathy ,Molecular Biology ,Genetics (clinical) ,Actin ,0303 health sciences ,Myocardium ,Body Weight ,Homozygote ,030305 genetics & heredity ,Intracellular Signaling Peptides and Proteins ,Cardiac muscle ,Skeletal muscle ,General Medicine ,LIM Domain Proteins ,medicine.disease ,Actins ,Muscular Dystrophy, Emery-Dreifuss ,FHL1 ,Phenotype ,Endocrinology ,medicine.anatomical_structure ,Echocardiography ,Body Composition ,Female ,General Article ,medicine.symptom ,Cardiomyopathies ,Signal Transduction - Abstract
X-linked scapuloperoneal myopathy (X-SM), one of Four-and-a-half LIM 1 (FHL1) related diseases, is an adult-onset slowly progressive myopathy, often associated with cardiomyopathy. We previously generated a knock-in mouse model that has the same mutation (c.365 G > C, p.W122S) as human X-SM patients. The mutant male mouse developed late-onset slowly progressive myopathy without cardiomyopathy. In this study, we observed that heterozygous (Het) and homozygous (Homo) female mice did not show alterations of skeletal muscle function or histology. In contrast, 20-month-old mutant female mice showed signs of cardiomyopathy on echocardiograms with increased systolic diameter [wild-type (WT): 2.74 ± 0.22 mm, mean ± standard deviation (SD); Het: 3.13 ± 0.11 mm, P
- Published
- 2018
35. Combined HDAC and Bromodomain Protein Inhibition Reprograms Tumor Cell Metabolism and Elicits Synthetic Lethality in Glioblastoma
- Author
-
Mike-Andrew Westhoff, Georg Karpel-Massler, Junfei Zhao, Chang Shu, Peter Canoll, Giulio Kleiner, Markus D. Siegelin, Maria J. Sanchez-Quintero, Yiru Zhang, Elena Bianchetti, Catarina M. Quinzii, Chiaki Tsuge Ishida, Wataru Ishida, and Sheng Fu L. Lo
- Subjects
0301 basic medicine ,Sorafenib ,Cancer Research ,Cell Survival ,Antineoplastic Agents ,Apoptosis ,Synthetic lethality ,Article ,Histone Deacetylases ,Transcriptome ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Panobinostat ,medicine ,Animals ,Humans ,Integrated stress response ,Cell Proliferation ,Cellular Reprogramming ,Xenograft Model Antitumor Assays ,Bromodomain ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Oncology ,chemistry ,Cancer cell ,Cancer research ,Histone deacetylase ,Glioblastoma ,Synthetic Lethal Mutations ,medicine.drug - Abstract
Purpose: Glioblastoma remains a challenge in oncology, in part due to tumor heterogeneity. Experimental Design: Patient-derived xenograft and stem-like glioblastoma cells were used as the primary model systems. Results: Based on a transcriptome and subsequent gene set enrichment analysis (GSEA), we show by using clinically validated compounds that the combination of histone deacetylase (HDAC) inhibition and bromodomain protein (BRD) inhibition results in pronounced synergistic reduction in cellular viability in patient-derived xenograft and stem-like glioblastoma cells. Transcriptome-based GSEA analysis suggests that metabolic reprogramming is involved with synergistic reduction of oxidative and glycolytic pathways in the combination treatment. Extracellular flux analysis confirms that combined HDAC inhibition and BRD inhibition blunts oxidative and glycolytic metabolism of cancer cells, leading to a depletion of intracellular ATP production and total ATP levels. In turn, energy deprivation drives an integrated stress response, originating from the endoplasmic reticulum. This results in an increase in proapoptotic Noxa. Aside from Noxa, we encounter a compensatory increase of antiapoptotic Mcl-1 protein. Pharmacologic, utilizing the FDA-approved drug sorafenib, and genetic inhibition of Mcl-1 enhanced the effects of the combination therapy. Finally, we show in orthotopic patient-derived xenografts of GBM, that the combination treatment reduces tumor growth, and that triple therapy involving the clinically validated compounds panobinostat, OTX015, and sorafenib further enhances these effects, culminating in a significant regression of tumors in vivo. Conclusions: Overall, these results warrant clinical testing of this novel, efficacious combination therapy. Clin Cancer Res; 24(16); 3941–54. ©2018 AACR.
- Published
- 2018
36. Emerging therapies for mitochondrial diseases
- Author
-
Valentina Emmanuele, Catarina M. Quinzii, and Michio Hirano
- Subjects
0301 basic medicine ,Mitochondrial DNA ,Mitochondrial Diseases ,Cell Transplantation ,Mitochondrial replacement therapy ,Genetic enhancement ,medicine.disease_cause ,Bioinformatics ,MELAS syndrome ,DNA, Mitochondrial ,Biochemistry ,Oxidative Phosphorylation ,Article ,03 medical and health sciences ,RNA, Transfer ,Mitophagy ,medicine ,Animals ,Humans ,Hypoxia ,Molecular Biology ,Clinical Trials as Topic ,Mutation ,business.industry ,Free Radical Scavengers ,Genetic Therapy ,medicine.disease ,Mitochondrial Replacement Therapy ,Heteroplasmy ,Exercise Therapy ,Mitochondria ,030104 developmental biology ,Mitochondrial biogenesis ,Dietary Supplements ,business - Abstract
For the vast majority of patients with mitochondrial diseases, only supportive and symptomatic therapies are available. However, in the last decade, due to extraordinary advances in defining the causes and pathomechanisms of these diverse disorders, new therapies are being developed in the laboratory and are entering human clinical trials. In this review, we highlight the current use of dietary supplement and exercise therapies as well as emerging therapies that may be broadly applicable across multiple mitochondrial diseases or tailored for specific disorders. Examples of non-tailored therapeutic targets include: activation of mitochondrial biogenesis, regulation of mitophagy and mitochondrial dynamics, bypass of biochemical defects, mitochondrial replacement therapy, and hypoxia. In contrast, tailored therapies are: scavenging of toxic compounds, deoxynucleoside and deoxynucleotide treatments, cell replacement therapies, gene therapy, shifting mitochondrial DNA mutation heteroplasmy, and stabilization of mutant mitochondrial transfer RNAs.
- Published
- 2018
37. Novel recessive mutations in COQ4 cause severe infantile cardiomyopathy and encephalopathy associated with CoQ10 deficiency
- Author
-
Marcello Ziosi, Catarina M. Quinzii, Neal Sondheimer, Jane Dunning Broadbent, Gino R. Somers, Jessie M. Cameron, Ali Naini, and Stacy Hewson
- Subjects
0301 basic medicine ,Infantile cardiomyopathy ,Mitochondrial disease ,Encephalopathy ,CoQ10 deficiency ,Encephalomyopathy ,Case Report ,03 medical and health sciences ,Endocrinology ,PDSS2 ,COQ6 ,Genetics ,COQ7 ,medicine ,Molecular Biology ,lcsh:QH301-705.5 ,lcsh:R5-920 ,business.industry ,Whole exome sequencing ,CoQ4 ,medicine.disease ,3. Good health ,030104 developmental biology ,Mitochondrial respiratory chain ,lcsh:Biology (General) ,Immunology ,Age of onset ,business ,lcsh:Medicine (General) ,ADCK3 - Abstract
Coenzyme Q10 (CoQ10) or ubiquinone is one of the two electron carriers in the mitochondrial respiratory chain which has an essential role in the process of oxidative phosphorylation. Defects in CoQ10 synthesis are usually associated with the impaired function of CoQ10–dependent complexes I, II and III. The recessively transmitted CoQ10 deficiency has been associated with a number of phenotypically and genetically heterogeneous groups of disorders manifesting at variable age of onset. The infantile, multisystemic presentation is usually caused by mutations in genes directly involved in CoQ10 biosynthesis. To date, mutations in COQ1 (PDSS1 and PDSS2), COQ2, COQ4, COQ6, COQ7, COQ8A/ADCK3, COQ8B/ADCK4, and COQ9 genes have been identified in patients with primary form of CoQ10 deficiency. Here we report novel mutations in the COQ4 gene, which were identified in an infant with profound mitochondrial disease presenting with perinatal seizures, hypertrophic cardiomyopathy and severe muscle CoQ10 deficiency.
- Published
- 2017
38. Loss of a novel striated muscle-enriched mitochondrial protein Coq10a enhances postnatal cardiac hypertrophic growth
- Author
-
Kazuki Tajima, Kentaro Hirose, Sheamin Khyeam, Jiajia Wang, Hongyao Yu, Guo N. Huang, Xiaoxin Chen, Steven Chang, Catarina M. Quinzii, Takeshi Yoneshiro, Shingo Kajimura, Emanuele Barca, and Guang Hu
- Subjects
chemistry.chemical_classification ,0303 health sciences ,Heart growth ,Mutant ,Peroxisome proliferator-activated receptor ,Peroxisome ,Biology ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,chemistry ,Mitochondrial biogenesis ,Coenzyme Q – cytochrome c reductase ,Coactivator ,Receptor ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Postnatal mammalian cardiomyocytes undergo a major transition from hyperplasia (increases in cell numbers) to hypertrophy (expansion in cell size). This process is accompanied by rapid mitochondrial biogenesis and metabolic switches to meet the demand of increased cardiac output. Although most mitochondrial components express ubiquitously, recent transcriptomic and proteomic analyses have discovered numerous tissue-specific mitochondrial proteins whose physiological functions are largely unknown. Here we report that a highly evolutionarily conserved mitochondrial protein Coq10a is predominantly expressed in mammalian cardiac and skeletal muscles, and is highly up-regulated around birth in a thyroid hormone-dependent manner. Deletion ofCoq10aby CRISPR/Cas9 leads to enhanced cardiac growth after birth. Surprisingly, adultCoq10amutant mice maintain the hypertrophic heart phenotype with increased levels of coenzyme Q (CoQ) per cardiomyocyte, preserved cardiac contractile function and mitochondrial respiration, which contrasts with reported mice and humans with mutations in other Coq family genes. Further RNA-seq analysis and mitochondrial characterization suggest an increase of mitochondrial biogenesis in theCoq10amutant heart as a possible consequence of Peroxisome proliferator-activated receptor Gamma Coactivator 1-alpha (PGC1α) activation, consistent with a recent intriguing report that CoQ may function as a natural ligand and partial agonist of Peroxisome Proliferator-Activated Receptor (PPAR) α/γ. Taken together, our study reveals a previously unknown function of a novel striated muscle-enriched mitochondrial protein Coq10a in regulating postnatal heart growth.
- Published
- 2019
39. Primary coenzyme Q10 deficiency-7: expanded phenotypic spectrum and a founder mutation in southern Chinese
- Author
-
Sophie Hon Yu Lai, Brian H.Y. Chung, Wuh-Liang Hwu, Wendy W.M. Lam, Shuk Ching Chong, Anna Ka Yee Kwong, Yen Yin Chou, Catarina M. Quinzii, Mandy H.Y. Tsang, Kit San Yeung, Jan A.M. Smeitink, Joannie Hui, Mullin H.C. Yu, Christopher C.Y. Mak, Jasmine L.F. Fung, Richard J. Rodenburg, Cheung Tsoi, Ni-Chung Lee, Pao Lin Kuo, Victor Chi Man Chan, Cheuk Wing Fung, Donald M.L. Tse, Shuan-Pei Lin, Brooke R. Willis, Matthew Ho, and Yin-Hsiu Chien
- Subjects
0301 basic medicine ,lcsh:QH426-470 ,Mitochondrial disease ,lcsh:Medicine ,Case Report ,Diseases ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,All institutes and research themes of the Radboud University Medical Center ,Genetics ,medicine ,Molecular Biology ,Founder mutation ,Genetics (clinical) ,Coenzyme Q10 ,Disease genetics ,lcsh:R ,Southern chinese ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,medicine.disease ,Phenotype ,lcsh:Genetics ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cohort ,Infantile onset ,Coenzyme Q10 deficiency - Abstract
Primary coenzyme Q10 deficiency-7 (COQ10D7) is a rare mitochondrial disease caused by biallelic mutations in COQ4. Here we report the largest cohort of COQ10D7 to date, with 11 southern Chinese patients confirmed with biallelic COQ4 mutations. Five of them have the classical neonatal-onset encephalo-cardiomyopathy, while the others have infantile onset with more heterogeneous clinical presentations. We also identify a founder mutation COQ4 (NM_016035.5): c.370G>A, p.(Gly124Ser) for COQ10D7, suggesting a higher chance of occurrence in the southern Chinese. This study helps improve understanding of the clinical spectrum of this disorder.
- Published
- 2019
40. Growth differentiation factor-15 as a biomarker of strength and recovery in survivors of acute respiratory failure
- Author
-
Catarina M. Quinzii, Michio Hirano, David J. Lederer, Matthew R. Baldwin, Elizabeth Colantuoni, Brian Rosenberg, and Dale M. Needham
- Subjects
Pulmonary and Respiratory Medicine ,Male ,medicine.medical_specialty ,Weakness ,Growth Differentiation Factor 15 ,medicine.medical_treatment ,Diaphragm ,Disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Mitochondrial myopathy ,Internal medicine ,Intensive care ,medicine ,Humans ,Pulmonary rehabilitation ,Survivors ,Aged ,Aged, 80 and over ,Hand Strength ,business.industry ,Recovery of Function ,Middle Aged ,medicine.disease ,Comorbidity ,Cross-Sectional Studies ,030228 respiratory system ,Acute Disease ,Biomarker (medicine) ,Female ,GDF15 ,medicine.symptom ,business ,Respiratory Insufficiency ,030217 neurology & neurosurgery ,Biomarkers - Abstract
Muscle mitochondrial dysfunction is implicated in intensive care unit-acquired weakness, but there is no serum biomarker of muscle mitochondrial function for critical illness survivors. Higher serum growth differentiation factor-15 (GDF-15) is a biomarker of inherited mitochondrial myopathy disease and is associated with mortality in several age-related diseases. Among 142 older (age ≥ 65 years) survivors of acute respiratory failure, we found that higher serum GDF-15 measured during the week prior to hospital discharge was cross-sectionally associated with weaker diaphragm, limb and hand-grip strength, and longitudinally associated with lower rates of functional recovery over 6 months, independent of age, sex, pre-existing disability, comorbidity, frailty, Acute Physiology and Chronic Health Evaluation II scores and concurrent interleukin-6 levels.
- Published
- 2019
41. MET Inhibition Elicits PGC1α-Dependent Metabolic Reprogramming in Glioblastoma
- Author
-
Catarina M. Quinzii, Junfei Zhao, Nelson Humala, Aayushi Mahajan, Chang Shu, Mike-Andrew Westhoff, Giulio Kleiner, Georg Karpel-Massler, Peter Canoll, Consuelo Torrini, Markus D. Siegelin, Maria J. Sanchez-Quintero, Yiru Zhang, Jeffrey N. Bruce, Angeliki Mela, Elena Bianchetti, Trang T. T. Nguyen, and Enyuan Shang
- Subjects
0301 basic medicine ,Proteomics ,Cancer Research ,Lactams, Macrocyclic ,Cell Respiration ,Oxidative phosphorylation ,Mitochondrion ,Guanidines ,Mitochondrial Dynamics ,Oxidative Phosphorylation ,Article ,Small hairpin RNA ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Crizotinib ,Carnitine ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Gene silencing ,Animals ,Humans ,Metabolomics ,Beta oxidation ,Cell Proliferation ,Kinase ,Brain Neoplasms ,Gene Expression Profiling ,Fatty Acids ,Drug Synergism ,Proto-Oncogene Proteins c-met ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Xenograft Model Antitumor Assays ,Mitochondria ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Epoxy Compounds ,Glioblastoma ,Reactive Oxygen Species ,Glycolysis ,Etomoxir ,medicine.drug - Abstract
The receptor kinase c-MET has emerged as a target for glioblastoma therapy. However, treatment resistance emerges inevitably. Here, we performed global metabolite screening with metabolite set enrichment coupled with transcriptome and gene set enrichment analysis and proteomic screening, and identified substantial reprogramming of tumor metabolism involving oxidative phosphorylation and fatty acid oxidation (FAO) with substantial accumulation of acyl-carnitines accompanied by an increase of PGC1α in response to genetic (shRNA and CRISPR/Cas9) and pharmacologic (crizotinib) inhibition of c-MET. Extracellular flux and carbon tracing analyses (U-13C-glucose, U-13C-glutamine, and U-13C-palmitic acid) demonstrated enhanced oxidative metabolism, which was driven by FAO and supported by increased anaplerosis of glucose carbons. These findings were observed in concert with increased number and fusion of mitochondria and production of reactive oxygen species. Genetic interference with PGC1α rescued this oxidative phenotype driven by c-MET inhibition. Silencing and chromatin immunoprecipitation experiments demonstrated that cAMP response elements binding protein regulates the expression of PGC1α in the context of c-MET inhibition. Interference with both oxidative phosphorylation (metformin, oligomycin) and β-oxidation of fatty acids (etomoxir) enhanced the antitumor efficacy of c-MET inhibition. Synergistic cell death was observed with c-MET inhibition and gamitrinib treatment. In patient-derived xenograft models, combination treatments of crizotinib and etomoxir, and crizotinib and gamitrinib were significantly more efficacious than single treatments and did not induce toxicity. Collectively, we have unraveled the mechanistic underpinnings of c-MET inhibition and identified novel combination therapies that may enhance its therapeutic efficacy. Significance: c-MET inhibition causes profound metabolic reprogramming that can be targeted by drug combination therapies.
- Published
- 2019
42. Serum Biomarkers of Mitochondrial Myopathy, Strength, and Recovery in Older Survivors of Acute Respiratory Failure
- Author
-
D.J. Lederer, Michio Hirano, Catarina M. Quinzii, Wendy C. Gonzalez, Matthew R. Baldwin, and Brian Rosenberg
- Subjects
medicine.medical_specialty ,Mitochondrial myopathy ,business.industry ,Serum biomarkers ,Internal medicine ,medicine ,Acute respiratory failure ,medicine.disease ,business ,Gastroenterology - Published
- 2019
43. Activation of LXRβ inhibits tumor respiration and is synthetically lethal with Bcl-xL inhibition
- Author
-
Giulio Kleiner, Chiaki Tsuge Ishida, Mike-Andrew Westhoff, Peter Canoll, Chang Shu, Trang T. T. Nguyen, Markus D. Siegelin, Maria J. Sanchez-Quintero, Yiru Zhang, Elena Bianchetti, Consuelo Torrini, Catarina M. Quinzii, Georg Karpel-Massler, and Enyuan Shang
- Subjects
0301 basic medicine ,BH3 mimetics ,Medicine (General) ,Benzylamines ,Apoptosis ,QH426-470 ,Benzoates ,Transcriptome ,0302 clinical medicine ,Melanoma ,Liver X Receptors ,Cancer ,biology ,Chemistry ,Articles ,3. Good health ,Treatment Outcome ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Programmed cell death ,Indazoles ,Cell Respiration ,bcl-X Protein ,Bcl-xL ,Article ,03 medical and health sciences ,R5-920 ,Chemical Biology ,Genetics ,medicine ,Extracellular ,Integrated stress response ,Animals ,Humans ,Metabolomics ,LXR agonist ,Liver X receptor ,Cell Proliferation ,Gene Expression Profiling ,Carcinoma ,electron transport chain ,glioblastoma ,Models, Theoretical ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Metabolism ,colon adenocarcinoma ,Cancer research ,biology.protein ,030217 neurology & neurosurgery - Abstract
Liver‐X‐receptor (LXR) agonists are known to bear anti‐tumor activity. However, their efficacy is limited and additional insights regarding the underlying mechanism are necessary. By performing transcriptome analysis coupled with global polar metabolite screening, we show that LXR agonists, LXR623 and GW3965, enhance synergistically the anti‐proliferative effect of BH3 mimetics in solid tumor malignancies, which is predominantly mediated by cell death with features of apoptosis and is rescued by exogenous cholesterol. Extracellular flux analysis and carbon tracing experiments (U‐13C‐glucose and U‐13C‐glutamine) reveal that within 5 h, activation of LXRβ results in reprogramming of tumor cell metabolism, leading to suppression of mitochondrial respiration, a phenomenon not observed in normal human astrocytes. LXR activation elicits a suppression of respiratory complexes at the protein level by reducing their stability. In turn, energy starvation drives an integrated stress response (ISR) that up‐regulates pro‐apoptotic Noxa in an ATF4‐dependent manner. Cholesterol and nucleotides rescue from the ISR elicited by LXR agonists and from cell death induced by LXR agonists and BH3 mimetics. In conventional and patient‐derived xenograft models of colon carcinoma, melanoma, and glioblastoma, the combination treatment of ABT263 and LXR agonists reduces tumor sizes significantly stronger than single treatments. Therefore, the combination treatment of LXR agonists and BH3 mimetics might be a viable efficacious treatment approach for solid malignancies.
- Published
- 2019
44. HDAC inhibitors elicit metabolic reprogramming by targeting super-enhancers in glioblastoma models
- Author
-
Georg Karpel-Massler, Yiru Zhang, Elena Bianchetti, Consuelo Torrini, Mike-Andrew Westhoff, Peter Canoll, Mark Maienschein-Cline, Markus D. Siegelin, Catarina M. Quinzii, Zhengdeng Lei, Enyuan Shang, Jeffrey N. Bruce, Junfei Zhao, Angeliki Mela, Nelson Humala, Trang T. T. Nguyen, Chang Shu, Arif Harmanci, and Aayushi Mahajan
- Subjects
0301 basic medicine ,Histone Deacetylase 2 ,Histone Deacetylase 1 ,Response Elements ,Oxidative Phosphorylation ,Romidepsin ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Panobinostat ,medicine ,Animals ,Humans ,Glycolysis ,PPAR delta ,Vorinostat ,Fatty Acids ,General Medicine ,Cellular Reprogramming ,HCT116 Cells ,Warburg effect ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,HDAC1 ,Histone Deacetylase Inhibitors ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Histone deacetylase ,Glioblastoma ,medicine.drug ,Research Article - Abstract
The Warburg effect is a tumor-related phenomenon that could potentially be targeted therapeutically. Here, we showed that glioblastoma (GBM) cultures and patients' tumors harbored super-enhancers in several genes related to the Warburg effect. By conducting a transcriptome analysis followed by ChIP-Seq coupled with a comprehensive metabolite analysis in GBM models, we found that FDA-approved global (panobinostat, vorinostat) and selective (romidepsin) histone deacetylase (HDAC) inhibitors elicited metabolic reprogramming in concert with disruption of several Warburg effect-related super-enhancers. Extracellular flux and carbon-tracing analyses revealed that HDAC inhibitors blunted glycolysis in a c-Myc-dependent manner and lowered ATP levels. This resulted in the engagement of oxidative phosphorylation (OXPHOS) driven by elevated fatty acid oxidation (FAO), rendering GBM cells dependent on these pathways. Mechanistically, interference with HDAC1/-2 elicited a suppression of c-Myc protein levels and a concomitant increase in 2 transcriptional drivers of oxidative metabolism, PGC1α and PPARD, suggesting an inverse relationship. Rescue and ChIP experiments indicated that c-Myc bound to the promoter regions of PGC1α and PPARD to counteract their upregulation driven by HDAC1/-2 inhibition. Finally, we demonstrated that combination treatment with HDAC and FAO inhibitors extended animal survival in patient-derived xenograft model systems in vivo more potently than single treatments in the absence of toxicity.
- Published
- 2019
45. A metabolic perspective on CSF-mediated neurodegeneration in multiple sclerosis
- Author
-
Hye-Jin Park, Matilde Inglese, Catarina M. Quinzii, Emily I. Chen, Vladimir Tolstikov, Patrizia Casaccia, Carlos Lopez-Gomez, Ilana Katz Sand, Valentina Fossati, James M. Aramini, Maureen Wentling, Tom Rusielewicz, Michael A. Kiebish, Mario Amatruda, Achilles Ntranos, and Maria Petracca
- Subjects
0301 basic medicine ,Hot Temperature ,Intravital Microscopy ,Glucose uptake ,Mitochondrion ,Relapsing-Remitting ,chemistry.chemical_compound ,0302 clinical medicine ,Phosphorylation ,Cerebrospinal Fluid ,Neurons ,Neurodegeneration ,Multiple Sclerosis, Chronic Progressive ,Mitochondria ,Chronic Progressive ,mitochondrial fusion ,Lactates ,metabolomic ,Dynamins ,medicine.medical_specialty ,Ceramide ,axonal damage ,Multiple Sclerosis ,Ceramides ,lipids ,03 medical and health sciences ,Multiple Sclerosis, Relapsing-Remitting ,Internal medicine ,medicine ,Animals ,Demyelinating Disorder ,Protein Processing ,business.industry ,Multiple sclerosis ,Post-Translational ,medicine.disease ,Rats ,030104 developmental biology ,Endocrinology ,Glucose ,chemistry ,clinical progression ,mitochondria ,Energy Metabolism ,Lipidomics ,Nerve Degeneration ,Protein Processing, Post-Translational ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Progressive disease - Abstract
Multiple sclerosis is an autoimmune demyelinating disorder of the CNS, characterized by inflammatory lesions and an underlying neurodegenerative process, which is more prominent in patients with progressive disease course. It has been proposed that mitochondrial dysfunction underlies neuronal damage, the precise mechanism by which this occurs remains uncertain. To investigate potential mechanisms of neurodegeneration, we conducted a functional screening of mitochondria in neurons exposed to the CSF of multiple sclerosis patients with a relapsing remitting (n = 15) or a progressive (secondary, n = 15 or primary, n = 14) disease course. Live-imaging of CSF-treated neurons, using a fluorescent mitochondrial tracer, identified mitochondrial elongation as a unique effect induced by the CSF from progressive patients. These morphological changes were associated with decreased activity of mitochondrial complexes I, III and IV and correlated with axonal damage. The effect of CSF treatment on the morphology of mitochondria was characterized by phosphorylation of serine 637 on the dynamin-related protein DRP1, a post-translational modification responsible for unopposed mitochondrial fusion in response to low glucose conditions. The effect of neuronal treatment with CSF from progressive patients was heat stable, thereby prompting us to conduct an unbiased exploratory lipidomic study that identified specific ceramide species as differentially abundant in the CSF of progressive patients compared to relapsing remitting multiple sclerosis. Treatment of neurons with medium supplemented with ceramides, induced a time-dependent increase of the transcripts levels of specific glucose and lactate transporters, which functionally resulted in progressively increased glucose uptake from the medium. Thus ceramide levels in the CSF of patients with progressive multiple sclerosis not only impaired mitochondrial respiration but also decreased the bioavailability of glucose by increasing its uptake. Importantly the neurotoxic effect of CSF treatment could be rescued by exogenous supplementation with glucose or lactate, presumably to compensate the inefficient fuel utilization. Together these data suggest a condition of ‘virtual hypoglycosis’ induced by the CSF of progressive patients in cultured neurons and suggest a critical temporal window of intervention for the rescue of the metabolic impairment of neuronal bioenergetics underlying neurodegeneration in multiple sclerosis patients.
- Published
- 2019
46. Anti-Oxidant Drugs: Novelties and Clinical Implications in Cerebellar Ataxias
- Author
-
Salvatore DiMauro, Valentina Emmanuele, Antonio Toscano, Catarina M. Quinzii, and Emanuele Barca
- Subjects
0301 basic medicine ,ataxias ,Disease ,Bioinformatics ,Article ,Antioxidants ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Idebenone ,Animals ,Humans ,oxidative stress ,Pharmacology (medical) ,Cerebellar ataxia ,Pharmacology ,business.industry ,Clinical course ,General Medicine ,Anti oxidant ,Bench to bedside ,Clinical trial ,Psychiatry and Mental health ,030104 developmental biology ,Cerebellar diseases ,Neurology ,Antioxidants, Cerebellar ataxia, Coenzyme Q10, Idebenone, ataxias, oxidative stress ,Coenzyme Q10 ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Background:Hereditary cerebellar ataxias are a group of disorders characterized by heterogeneous clinical manifestations, progressive clinical course, and diverse genetic causes. No disease modifying treatments are yet available for many of these disorders. Oxidative stress has been recurrently identified in different progressive cerebellar diseases, and it represents a widely investigated target for treatment. Objective: To review the main aspects and new perspectives of antioxidant therapy in cerebellar ataxias ranging from bench to bedside. Method: This article is a summary of the state-of-the-art on the use of antioxidant molecules in cerebellar ataxia treatments. It also briefly summarizes aspects of oxidative stress production and general characteristics of antioxidant compounds. Results: Antioxidants represent a vast category of compounds; old drugs have been extensively studied and modified in order to achieve better biological effects. Despite the vast body of literature present on the use of antioxidants in cerebellar ataxias, for the majority of these disorders conclusive results on the efficacy are still missing.Conclusion:Antioxidant therapy in cerebellar ataxias is a promising field of investigations. To achieve the success in identifying the correct treatment more work needs to be done. In particular, a combined effort is needed by basic scientists in developing more efficient molecules, and by clinical researchers together with patients communities, to run clinical trials in order to identify conclusive treatments strategies.
- Published
- 2019
47. Coenzyme Q10 Deficiency
- Author
-
Luis C. López and Catarina M. Quinzii
- Subjects
Coenzyme Q10 ,medicine.medical_specialty ,Cerebellar ataxia ,Chemistry ,medicine.disease_cause ,medicine.disease ,Nephropathy ,chemistry.chemical_compound ,Endocrinology ,Internal medicine ,Coenzyme Q – cytochrome c reductase ,medicine ,Lipid molecule ,medicine.symptom ,Myopathy ,Oxidative stress - Abstract
Coenzyme Q10 (CoQ10) deficiency is defined as the low amount of the lipid molecule CoQ10 in muscle and/or other tissue.
- Published
- 2019
48. Metabolic Targets of Coenzyme Q10 in Mitochondria
- Author
-
Catarina M. Quinzii, Luis C. López, María E. Díaz-Casado, Sergio López-Herrador, Eliana Barriocanal-Casado, Pilar González-García, and Agustín Hidalgo-Gutiérrez
- Subjects
0301 basic medicine ,Physiology ,sulfide metabolism ,Clinical Biochemistry ,Review ,Oxidative phosphorylation ,super-complexes ,Mitochondrion ,ubiquinol-10 ,Ubiquinone-10 ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,ubiquinone-10 ,0302 clinical medicine ,Biosynthesis ,coenzyme Q10 ,proline metabolism ,Ubiquinol-10 ,Molecular Biology ,Coenzyme Q10 ,Sulfide metabolism ,Catabolism ,lcsh:RM1-950 ,food and beverages ,OxPhos ,Cell Biology ,Metabolism ,one-carbon metabolism ,One-carbon metabolism ,Mitochondria ,mitochondria ,Metabolic pathway ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,Mitochondrial respiratory chain ,chemistry ,Super-complexes ,Proline metabolism ,030217 neurology & neurosurgery - Abstract
This work was supported by grants from Ministerio de Ciencia e Innovacion, Spain, and the ERDF (RTI2018-093503-B-100), the Muscular Dystrophy Association (MDA-602322). C.M.Q. is supported by the Department of Defense (DOD) grant PR190511. A.H.-G. and P.G.-G. are `FPU fellows' from the Ministerio de Universidades, Spain. S.L.-H. is supported by the "becas de colaboracion" from the Ministerio de Universidades, Spain. E.B.-C. is supported by the Consejeria de Salud, Junta de Andalucia, Spain., We thank Stacy Kelly Aguirre for the English editing. Figures created with BioRender.com., Coenzyme Q10 (CoQ(10)) is classically viewed as an important endogenous antioxidant and key component of the mitochondrial respiratory chain. For this second function, CoQ molecules seem to be dynamically segmented in a pool attached and engulfed by the super-complexes I + III, and a free pool available for complex II or any other mitochondrial enzyme that uses CoQ as a cofactor. This CoQ-free pool is, therefore, used by enzymes that link the mitochondrial respiratory chain to other pathways, such as the pyrimidine de novo biosynthesis, fatty acid beta-oxidation and amino acid catabolism, glycine metabolism, proline, glyoxylate and arginine metabolism, and sulfide oxidation metabolism. Some of these mitochondrial pathways are also connected to metabolic pathways in other compartments of the cell and, consequently, CoQ could indirectly modulate metabolic pathways located outside the mitochondria. Thus, we review the most relevant findings in all these metabolic functions of CoQ and their relations with the pathomechanisms of some metabolic diseases, highlighting some future perspectives and potential therapeutic implications., Spanish Government, European Commission RTI2018-093503-B-100, Muscular Dystrophy Association MDA-602322, United States Department of Defense PR190511, Ministerio de Universidades, Spain, Junta de Andalucia
- Published
- 2021
49. ETMM-04. AURKA INHIBITION REPROGRAMS METABOLISM AND IS SYNTHETICALLY LETHAL WITH FATTY ACID OXIDATION INHIBITION IN GLIOBLASTOMA MODEL SYSTEMS
- Author
-
Mike-Andrew Westhof, Peter Canoll, Jeffrey N. Bruce, Nelson Humala, Aayushi Mahajan, Markus D. Siegelin, Angeliki Mela, Chang Shu, Trang T. T. Nguyen, Georg Karpel-Massler, Catarina M. Quinzii, Hasan O. Akman, Guoan Zhang, and Enyuan Shang
- Subjects
Chromatin ,Cell biology ,Supplement Abstracts ,Small hairpin RNA ,chemistry.chemical_compound ,chemistry ,Epigenome, Transcriptome, Metabolome and Modeling ,Alisertib ,Gene silencing ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Aurora Kinase A ,Growth inhibition ,Signal transduction ,Chromatin immunoprecipitation - Abstract
Aurora kinase A (AURKA) has emerged as a viable drug target for glioblastoma (GBM), the most common malignant primary brain tumor in adults with a life expectancy of 12–15 months. However, resistance to therapy remains a critical issue, which partially may be driven by reprogramming of metabolism. By integration of transcriptome, chromatin immunoprecipitation with sequencing (CHIP-seq.), assay for transposase-accessible chromatin with sequencing (ATAC-seq.), proteomic and metabolite screening followed by carbon tracing (U-13C-Glucose, U-13C-Glutamine and U-13C-Palmitic acid) and extracellular flux analysis we provided evidence that genetic (shRNA and CRISPR/Cas9) and pharmacological (Alisertib) AURKA inhibition elicited substantial metabolic reprogramming supported in part by inhibition of MYC targets and concomitant activation of PPARA signaling. While glycolysis was suppressed by AURKA inhibition, we noted a compensatory increase in oxygen consumption rate fueled by enhanced fatty acid oxidation (FAO). Whereas interference with AURKA elicited a suppression of c-Myc, we detected an upregulation of PGC1A, a master regulator of oxidative metabolism. Silencing of PGC1A reversed AURKAi mediated metabolic reprogramming and sensitized GBM cells to AURKAi driven reduction of cellular viability. Chromatin immunoprecipitation experiments showed binding of c-Myc to the promoter region of PGC1A, which is abrogated by AURKA inhibition and in turn unleashed PGC1A expression. Consistently, ATAC-seq. confirmed higher accessibility of a MYC binding region within the PGC1A promoter, suggesting that MYC acts as a repressor of PGC1A. Combining alisertib with inhibitors of FAO or the electron transport chain exerted substantial synergistic growth inhibition in PDX lines in vitro and extension of overall survival in orthotopic GBM PDX models without induction of toxicity in normal tissue. In summary, these findings support that simultaneous targeting of oxidative energy metabolism and AURKAi might be a potential novel therapy against GBM.
- Published
- 2021
50. Decreased Coenzyme Q10 Levels in Multiple System Atrophy Cerebellum
- Author
-
Emanuele Barca, Elan D. Louis, Guomei Tang, Marcello Ziosi, Jose L Torres, Eliezer Masliah, Giulio Kleiner, Un Jung Kang, Jean Paul G. Vonsattel, Sheng-Han Kuo, Etty Cortes, Phyllis L. Faust, Catarina M. Quinzii, and Saba Tadesse
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Cerebellum ,Mitochondrial Diseases ,Ataxia ,Ubiquinone ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Pathogenesis ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Atrophy ,Internal medicine ,medicine ,Humans ,Cerebellar ataxia ,Aged ,Aged, 80 and over ,Coenzyme Q10 ,Muscle Weakness ,Original Articles ,Multiple system atrophy ,General Medicine ,Middle Aged ,medicine.disease ,030104 developmental biology ,Endocrinology ,Mitochondrial respiratory chain ,medicine.anatomical_structure ,Neurology ,chemistry ,Oxidative stress ,Case-Control Studies ,Female ,Neurology (clinical) ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
In familial and sporadic multiple system atrophy (MSA) patients, deficiency of coenzyme Q10 (CoQ10) has been associated with mutations in COQ2, which encodes the second enzyme in the CoQ10 biosynthetic pathway. Cerebellar ataxia is the most common presentation of CoQ10 deficiency, suggesting that the cerebellum might be selectively vulnerable to low levels of CoQ10. To investigate whether CoQ10 deficiency represents a common feature in the brains of MSA patients independent of the presence of COQ2 mutations, we studied CoQ10 levels in postmortem brains of 12 MSA, 9 Parkinson disease (PD), 9 essential tremor (ET) patients, and 12 controls. We also assessed mitochondrial respiratory chain enzyme activities, oxidative stress, mitochondrial mass, and levels of enzymes involved in CoQ biosynthesis. Our studies revealed CoQ10 deficiency in MSA cerebellum, which was associated with impaired CoQ biosynthesis and increased oxidative stress in the absence of COQ2 mutations. The levels of CoQ10 in the cerebella of ET and PD patients were comparable or higher than in controls. These findings suggest that CoQ10 deficiency may contribute to the pathogenesis of MSA. Because no disease modifying therapies are currently available, increasing CoQ10 levels by supplementation or upregulation of its biosynthesis may represent a novel treatment strategy for MSA patients.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.