185 results on '"Avadhani NG"'
Search Results
2. Genetic transformation in Escherichia coli
- Author
-
Rege Dv, Mehta Bm, and Avadhani Ng
- Subjects
Genetics, Microbial ,Lysis ,Auxotrophy ,Mutant ,Biology ,medicine.disease_cause ,chemistry.chemical_compound ,Methionine ,Transformation, Genetic ,Structural Biology ,medicine ,Escherichia coli ,Molecular Biology ,Homocysteine ,Temperature ,Biological activity ,DNA ,Hydrogen-Ion Concentration ,Transformation (genetics) ,Kinetics ,Vitamin B 12 ,Biochemistry ,chemistry - Abstract
The earlier observation of a DNA-induced genetic change in the cells of an auxotrophic strain Escherichia coli 113-3 dependent on vitamin B 12 or methionine for growth has been confirmed and extended, and a method for the isolation of biologically active DNA is described. Optimum conditions for transformation have been determined with respect to the pH of the cell lysate during extraction, concentration of DNA in the system and the temperature at which the competent cells are exposed to DNA. It is shown that the competence of the cells reaches a peak at the middle of the logarithmic phase of growth. A comparison of vitamin B 12 content and methionine-synthesizing abilities of the donor, recipient and transformed strains has confirmed the rehabilitation of the mutants to prototrophy. This observation has been extended to other strains of E. coli as donors and acceptors.
- Published
- 1969
3. Alcohol-induced CYP2E1, mitochondrial dynamics and retrograde signaling in human hepatic 3D organoids.
- Author
-
Angireddy R, Chowdhury AR, Zielonka J, Ruthel G, Kalyanaraman B, and Avadhani NG
- Subjects
- Humans, Liver metabolism, Organoids metabolism, Oxidative Stress, Cytochrome P-450 CYP2E1 genetics, Cytochrome P-450 CYP2E1 metabolism, Mitochondrial Dynamics
- Abstract
Alcohol toxicity is a significant health problem with ~3 million estimated deaths per year globally. Alcohol is metabolized to the toxic metabolite, acetaldehyde by alcohol dehydrogenase or CYP2E1 in the hepatic tissue, and also induces reactive oxygen species (ROS), which together play a pivotal role in cell and tissue damage. Our previous studies with COS-7 cells transduced with unique human CYP2E1 variants that mostly localize to either microsomes or mitochondria revealed that mitochondrially-localized CYP2E1 drives alcohol toxicity through the generation of higher levels of ROS, which has a consequent effect on cytochrome c oxidase (CcO) and mitochondrial oxidative function. Alcohol treatment of human hepatocyte cell line, HepaRG, in monolayer cultures increased ROS, affected CcO activity/stability, and induced mitophagy. Alcohol treatment of 3D organoids of HepaRG cells induced higher levels of CYP2E1 mRNA and activated mitochondrial stress-induced retrograde signaling, and also induced markers of hepatic steatosis. Knock down of CYP2E1 mRNA using specific shRNA, FK506, a Calcineurin inhibitor, and Mdivi-1, a DRP1 inhibitor, ameliorated alcohol-induced mitochondrial retrograde signaling, and hepatic steatosis. These results for the first time present a mechanistic link between CYP2E1 function and alcohol mediated mitochondrial dysfunction, retrograde signaling, and activation of hepatic steatosis in a 3D organoid system that closely recapitulates the in vivo liver response., Competing Interests: Declaration of competing interest The authors declare that they have no conflict of interest with the contents of this article., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
4. Mitochondria-targeted paraquat and metformin mediate ROS production to induce multiple pathways of retrograde signaling: A dose-dependent phenomenon.
- Author
-
Chowdhury AR, Zielonka J, Kalyanaraman B, Hartley RC, Murphy MP, and Avadhani NG
- Subjects
- Hydrogen Peroxide, Mitochondria, Reactive Oxygen Species, Signal Transduction, Metformin pharmacology, Paraquat
- Abstract
The mitochondrial electron transport chain is a major source of reactive oxygen species (ROS) and is also a target of ROS, with an implied role in the stabilization of hypoxia-inducible factor (HIF) and induction of the AMPK pathway. Here we used varying doses of two agents, Mito-Paraquat and Mito-Metformin, that have been conjugated to cationic triphenylphosphonium (TPP
+ ) moiety to selectively target them to the mitochondrial matrix compartment, thereby resulting in the site-specific generation of ROS within mitochondria. These agents primarily induce superoxide (O2 •- ) production by acting on complex I. In Raw264.7 macrophages, C2C12 skeletal myocytes, and HCT116 adenocarcinoma cells, we show that mitochondria-targeted oxidants can induce ROS (O2 •- and H2 O2 ). In all three cell lines tested, the mitochondria-targeted agents disrupted membrane potential and activated calcineurin and the Cn-dependent retrograde signaling pathway. Hypoxic culture conditions also induced Cn activation and HIF1α activation in a temporally regulated manner, with the former appearing at shorter exposure times. Together, our results indicate that mitochondrial oxidant-induced retrograde signaling is driven by disruption of membrane potential and activation of Ca2+ /Cn pathway and is independent of ROS-induced HIF1α or AMPK pathways., (Copyright © 2020. Published by Elsevier B.V.)- Published
- 2020
- Full Text
- View/download PDF
5. Dysregulation of RyR Calcium Channel Causes the Onset of Mitochondrial Retrograde Signaling.
- Author
-
Roy Chowdhury A, Srinivasan S, Csordás G, Hajnóczky G, and Avadhani NG
- Abstract
This study shows that multiple modes of mitochondrial stress generated by partial mtDNA depletion or cytochrome c oxidase disruption cause ryanodine receptor channel (RyR) dysregulation, which instigates the release of Ca
2+ in the cytoplasm of C2C12 myoblasts and HCT116 carcinoma cells. We also observed a reciprocal downregulation of IP3R channel activity and reduced mitochondrial uptake of Ca2+ . Ryanodine, an RyR antagonist, abrogated the mitochondrial stress-mediated increase in [Ca2+ ]c and the entire downstream signaling cascades of mitochondrial retrograde signaling. Interestingly, ryanodine also inhibited mitochondrial stress-induced invasive behavior in mtDNA-depleted C2C12 cells and HCT116 carcinoma cells. In addition, co-immunoprecipitation shows reduced FKBP12 protein binding to RyR channel proteins, suggesting the altered function of the Ca2+ channel. These results document how the endoplasmic reticulum-associated RyR channels, in combination with inhibition of the mitochondrial uniporter system, modulate cellular Ca2+ homeostasis and signaling under mitochondrial stress conditions., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
6. YY1 control of mitochondrial-related genes does not account for regulation of immunoglobulin class switch recombination in mice.
- Author
-
Nandi S, Liang G, Sindhava V, Angireddy R, Basu A, Banerjee S, Hodawadekar S, Zhang Y, Avadhani NG, Sen R, and Atchison ML
- Subjects
- Animals, B-Lymphocytes cytology, DNA, Mitochondrial genetics, Mice, Mice, Knockout, Spleen cytology, YY1 Transcription Factor genetics, B-Lymphocytes immunology, DNA, Mitochondrial immunology, Genes, Mitochondrial immunology, Immunoglobulin Class Switching, Spleen immunology, YY1 Transcription Factor immunology
- Abstract
Immunoglobulin class switch recombination (CSR) occurs in activated B cells with increased mitochondrial mass and membrane potential. Transcription factor Yin Yang 1 (YY1) is critical for CSR and for formation of the DNA loops involved in this process. We therefore sought to determine if YY1 knockout impacts mitochondrial gene expression and mitochondrial function in murine splenic B cells, providing a potential mechanism for regulating CSR. We identified numerous genes in splenic B cells differentially regulated when cells are induced to undergo CSR. YY1 conditional knockout caused differential expression of 1129 genes, with 59 being mitochondrial-related genes. ChIP-seq analyses showed YY1 was directly bound to nearly half of these mitochondrial-related genes. Surprisingly, at the time when YY1 knockout dramatically reduces DNA loop formation and CSR, mitochondrial mass and membrane potential were not significantly impacted, nor was there a significant change in mitochondrial oxygen consumption, extracellular acidification rate, or mitochondrial complex I or IV activities. Our results indicate that YY1 regulates numerous mitochondrial-related genes in splenic B cells, but this does not account for the impact of YY1 on CSR or long-distance DNA loop formation., (© 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2020
- Full Text
- View/download PDF
7. Esophageal 3D organoids of MPV17 -/- mouse model of mitochondrial DNA depletion show epithelial cell plasticity and telomere attrition.
- Author
-
Guha M, Srinivasan S, Sheehan MM, Kijima T, Ruthel G, Whelan K, Tanaka K, Klein-Szanto A, Chandramouleeswaran PM, Nakagawa H, and Avadhani NG
- Abstract
Esophageal squamous cell carcinoma (ESCC) is an aggressive cancer with late-stage detection and poor prognosis. This emphasizes the need to identify new markers for early diagnosis and treatment. Altered mitochondrial genome (mtDNA) content in primary tumors correlates with poor patient prognosis. Here we used three-dimensional (3D) organoids of esophageal epithelial cells (EECs) from the MPV17
-/- mouse model of mtDNA depletion to investigate the contribution of reduced mtDNA content in ESCC oncogenicity. To test if mtDNA defects are a contributing factor in ESCC, we used oncogenic stimuli such as ESCC carcinogen 4-nitroquinoline oxide (4-NQO) treatment, or expressing p53R175H oncogenic driver mutation. We observed that EECs and 3D-organoids with mtDNA depletion had cellular, morphological and genetic alterations typical of an oncogenic transition. Furthermore, mitochondrial dysfunction induced cellular transformation is accompanied by elevated mitochondrial fission protein, DRP1 and pharmacologic inhibition of mitochondrial fission by mDivi-1 in the MPV17-/- organoids reversed the phenotype to that of normal EEC organoids. Our studies show that mtDNA copy number depletion, activates a mitochondrial retrograde response, potentiates telomere defects, and increases the oncogenic susceptibility towards ESCC. Furthermore, mtDNA depletion driven cellular plasticity is mediated via altered mitochondrial fission-fusion dynamics., Competing Interests: CONFLICTS OF INTEREST The authors declare no conflicts of interest with this research. The Authors have no conflicts of interest with data presented in this study.- Published
- 2019
- Full Text
- View/download PDF
8. Cytochrome c oxidase dysfunction enhances phagocytic function and osteoclast formation in macrophages.
- Author
-
Angireddy R, Kazmi HR, Srinivasan S, Sun L, Iqbal J, Fuchs SY, Guha M, Kijima T, Yuen T, Zaidi M, and Avadhani NG
- Subjects
- Animals, Cell Differentiation, Electron Transport Complex IV antagonists & inhibitors, Electron Transport Complex IV genetics, Gene Knockdown Techniques, Macrophages classification, Membrane Proteins deficiency, Membrane Proteins genetics, Mice, Mice, Inbred BALB C, Mice, Knockout, Mitochondria metabolism, Osteogenesis, RAW 264.7 Cells, RNA, Small Interfering genetics, Reactive Oxygen Species metabolism, Signal Transduction, Stress, Physiological, Electron Transport Complex IV metabolism, Macrophages cytology, Macrophages physiology, Osteoclasts cytology, Osteoclasts physiology, Phagocytosis physiology
- Abstract
The mitochondria-to-nucleus retrograde signaling (MtRS) pathway aids in cellular adaptation to stress. We earlier reported that the Ca
2+ - and calcineurin-dependent MtRS induces macrophage differentiation to bone-resorbing osteoclasts. However, mechanisms through which macrophages sense and respond to cellular stress remain unclear. Here, we induced mitochondrial stress in macrophages by knockdown (KD) of subunits IVi1 or Vb of cytochrome c oxidase (CcO). Whereas both IVi1 and Vb KD impair CcO activity, IVi1 KD cells produced higher levels of cellular and mitochondrial reactive oxygen species with increased glycolysis. Additionally, IVi1 KD induced the activation of MtRS factors NF-κB, NFAT2, and C/EBPδ as well as inflammatory cytokines, NOS 2, increased phagocytic activity, and a greater osteoclast differentiation potential at suboptimal RANK-L concentrations. The osteoclastogenesis in IVi1 KD cells was reversed fully with an IL-6 inhibitor LMT-28, whereas there was minimal rescue of the enhanced phagocytosis in these cells. In agreement with our findings in cultured macrophages, primary bone marrow-derived macrophages from MPV17-/- mice, a model for mitochondrial dysfunction, also showed higher propensity for osteoclast formation. This is the first report showing that CcO dysfunction affects inflammatory pathways, phagocytic function, and osteoclastogenesis.-Angireddy, R., Kazmi, H. R., Srinivasan, S., Sun, L., Iqbal, J., Fuchs, S. Y., Guha, M., Kijima, T., Yuen, T., Zaidi, M., Avadhani, N. G. Cytochrome c oxidase dysfunction enhances phagocytic function and osteoclast formation in macrophages.- Published
- 2019
- Full Text
- View/download PDF
9. Mitochondrially targeted cytochrome P450 2D6 is involved in monomethylamine-induced neuronal damage in mouse models.
- Author
-
Chattopadhyay M, Chowdhury AR, Feng T, Assenmacher CA, Radaelli E, Guengerich FP, and Avadhani NG
- Subjects
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine adverse effects, Animals, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mitochondria drug effects, Mitochondria metabolism, Neuroblastoma etiology, Neuroblastoma metabolism, Neurons drug effects, Neurons metabolism, Neurotoxins toxicity, Parkinson Disease etiology, Parkinson Disease metabolism, Tumor Cells, Cultured, Cytochrome P-450 CYP2D6 physiology, Disease Models, Animal, Methylamines toxicity, Mitochondria pathology, Neuroblastoma pathology, Neurons pathology, Parkinson Disease pathology
- Abstract
Parkinson's disease (PD) is a major human disease associated with degeneration of the central nervous system. Evidence suggests that several endogenously formed 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mimicking chemicals that are metabolic conversion products, especially β-carbolines and isoquinolines, act as neurotoxins that induce PD or enhance progression of the disease. We have demonstrated previously that mitochondrially targeted human cytochrome P450 2D6 (CYP2D6), supported by mitochondrial adrenodoxin and adrenodoxin reductase, can efficiently catalyze the conversion of MPTP to the toxic 1-methyl-4-phenylpyridinium ion. In this study, we show that the mitochondrially targeted CYP2D6 can efficiently catalyze MPTP-mimicking compounds, i.e. 2-methyl-1,2,3,4-tetrahydroisoquinoline, 2-methyl-1,2,3,4-tetrahydro-β-carboline, and 9-methyl-norharmon, suspected to induce PD in humans. Our results reveal that activity and respiration in mouse brain mitochondrial complex I are significantly affected by these toxins in WT mice but remain unchanged in Cyp2d6 locus knockout mice, indicating a possible role of CYP2D6 in the metabolism of these compounds both in vivo and in vitro These metabolic effects were minimized in the presence of two CYP2D6 inhibitors, quinidine and ajmalicine. Neuro-2a cells stably expressing predominantly mitochondrially targeted CYP2D6 were more sensitive to toxin-mediated respiratory dysfunction and complex I inhibition than cells expressing predominantly endoplasmic reticulum-targeted CYP2D6. Exposure to these toxins also induced the autophagic marker Parkin and the mitochondrial fission marker Dynamin-related protein 1 (Drp1) in differentiated neurons expressing mitochondrial CYP2D6. Our results show that monomethylamines are converted to their toxic cationic form by mitochondrially directed CYP2D6 and result in neuronal degradation in mice., (© 2019 Chattopadhyay et al.)
- Published
- 2019
- Full Text
- View/download PDF
10. Correction to: HnRNPA2 is a novel histone acetyltransferase that mediates mitochondrial stress-induced nuclear gene expression.
- Author
-
Guha M, Srinivasan S, Guja K, Mejia E, Garcia-Diaz M, Johnson FB, Ruthel G, Kaufman BA, Rappaport EF, Glineburg MR, Fang JK, Klein-Szanto AJ, Nakagawa H, Basha J, Kundu T, and Avadhani NG
- Abstract
[This corrects the article DOI: 10.1038/celldisc.2016.45.].
- Published
- 2019
- Full Text
- View/download PDF
11. Cigarette Smoke Toxins-Induced Mitochondrial Dysfunction and Pancreatitis Involves Aryl Hydrocarbon Receptor Mediated Cyp1 Gene Expression: Protective Effects of Resveratrol.
- Author
-
Ghosh J, Chowdhury AR, Srinivasan S, Chattopadhyay M, Bose M, Bhattacharya S, Raza H, Fuchs SY, Rustgi AK, Gonzalez FJ, and Avadhani NG
- Subjects
- Animals, Cytokines metabolism, Female, Male, Mice, Mice, Knockout, Pancreatitis physiopathology, Smoke adverse effects, Nicotiana adverse effects, Benzo(a)pyrene toxicity, Cytochrome P450 Family 1 metabolism, Mitochondria drug effects, Mitochondria metabolism, Pancreatitis chemically induced, Polychlorinated Dibenzodioxins toxicity, Receptors, Aryl Hydrocarbon metabolism, Resveratrol pharmacology
- Abstract
We previously reported that mitochondrial CYP1 enzymes participate in the metabolism of polycyclic aromatic hydrocarbons and other carcinogens leading to mitochondrial dysfunction. In this study, using Cyp1b1-/-, Cyp1a1/1a2-/-, and Cyp1a1/1a2/1b1-/- mice, we observed that cigarette and environmental toxins, namely benzo[a]pyrene (BaP) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), induce pancreatic mitochondrial respiratory dysfunction and pancreatitis. Our results suggest that aryl hydrocarbon receptor (AhR) activation and resultant mitochondrial dysfunction are associated with pancreatic pathology. BaP treatment markedly inhibits pancreatic mitochondrial oxygen consumption rate (OCR), ADP-dependent OCR, and also maximal respiration, in wild-type mice but not in Cyp1a1/1a2-/- and Cyp1a1/1a2/1b1-/- mice. In addition, both BaP and TCDD treatment markedly affected mitochondrial complex IV activity, in addition to causing marked reduction in mitochondrial DNA content. Interestingly, the AhR antagonist resveratrol, attenuated BaP-induced mitochondrial respiratory defects in the pancreas, and reversed pancreatitis, both histologically and biochemically in wild-type mice. These results reveal a novel role for AhR- and AhR-regulated CYP1 enzymes in eliciting mitochondrial dysfunction and cigarette toxin-mediated pancreatic pathology. We propose that increased mitochondrial respiratory dysfunction and oxidative stress are involved in polycyclic aromatic hydrocarbon associated pancreatitis. Resveratrol, a chemo preventive agent and AhR antagonist, and CH-223191, a potent and specific AhR inhibitor, confer protection against BaP-induced mitochondrial dysfunction and pancreatic pathology.
- Published
- 2018
- Full Text
- View/download PDF
12. hnRNPA2 mediated acetylation reduces telomere length in response to mitochondrial dysfunction.
- Author
-
Guha M, Srinivasan S, Johnson FB, Ruthel G, Guja K, Garcia-Diaz M, Kaufman BA, Glineburg MR, Fang J, Nakagawa H, Basha J, Kundu T, and Avadhani NG
- Subjects
- Acetylation, Animals, Cell Line, Cell Transformation, Neoplastic genetics, Chromosomal Instability physiology, Epigenesis, Genetic physiology, Fibroblasts, Heterogeneous-Nuclear Ribonucleoprotein Group A-B genetics, Humans, Lysine metabolism, Mice, Mice, Inbred BALB C, Mice, Knockout, Mutagenesis, Site-Directed, Mutation, Telomerase metabolism, Heterogeneous-Nuclear Ribonucleoprotein Group A-B metabolism, Histones metabolism, Mitochondria metabolism, Telomere metabolism, Telomere Shortening genetics
- Abstract
Telomeres protect against chromosomal damage. Accelerated telomere loss has been associated with premature aging syndromes such as Werner's syndrome and Dyskeratosis Congenita, while, progressive telomere loss activates a DNA damage response leading to chromosomal instability, typically observed in cancer cells and senescent cells. Therefore, identifying mechanisms of telomere length maintenance is critical for understanding human pathologies. In this paper we demonstrate that mitochondrial dysfunction plays a causal role in telomere shortening. Furthermore, hnRNPA2, a mitochondrial stress responsive lysine acetyltransferase (KAT) acetylates telomere histone H4at lysine 8 of (H4K8) and this acetylation is associated with telomere attrition. Cells containing dysfunctional mitochondria have higher telomere H4K8 acetylation and shorter telomeres independent of cell proliferation rates. Ectopic expression of KAT mutant hnRNPA2 rescued telomere length possibly due to impaired H4K8 acetylation coupled with inability to activate telomerase expression. The phenotypic outcome of telomere shortening in immortalized cells included chromosomal instability (end-fusions) and telomerase activation, typical of an oncogenic transformation; while in non-telomerase expressing fibroblasts, mitochondrial dysfunction induced-telomere attrition resulted in senescence. Our findings provide a mechanistic association between dysfunctional mitochondria and telomere loss and therefore describe a novel epigenetic signal for telomere length maintenance., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
13. Three-Dimensional Organoids Reveal Therapy Resistance of Esophageal and Oropharyngeal Squamous Cell Carcinoma Cells.
- Author
-
Kijima T, Nakagawa H, Shimonosono M, Chandramouleeswaran PM, Hara T, Sahu V, Kasagi Y, Kikuchi O, Tanaka K, Giroux V, Muir AB, Whelan KA, Ohashi S, Naganuma S, Klein-Szanto AJ, Shinden Y, Sasaki K, Omoto I, Kita Y, Muto M, Bass AJ, Diehl JA, Ginsberg GG, Doki Y, Mori M, Uchikado Y, Arigami T, Avadhani NG, Basu D, Rustgi AK, and Natsugoe S
- Subjects
- Animals, Autophagy drug effects, Biopsy, Carcinoma, Squamous Cell therapy, Cell Line, Tumor, Chemoradiotherapy, Endoscopy, Fluorouracil pharmacology, Fluorouracil therapeutic use, Humans, Hyaluronan Receptors metabolism, Mice, Oropharyngeal Neoplasms therapy, Carcinoma, Squamous Cell pathology, Drug Resistance, Neoplasm, Esophageal Neoplasms pathology, Organoids pathology, Oropharyngeal Neoplasms pathology
- Abstract
Background & Aims: Oropharyngeal and esophageal squamous cell carcinomas, especially the latter, are a lethal disease, featuring intratumoral cancer cell heterogeneity and therapy resistance. To facilitate cancer therapy in personalized medicine, three-dimensional (3D) organoids may be useful for functional characterization of cancer cells ex vivo . We investigated the feasibility and the utility of patient-derived 3D organoids of esophageal and oropharyngeal squamous cell carcinomas., Methods: We generated 3D organoids from paired biopsies representing tumors and adjacent normal mucosa from therapy-naïve patients and cell lines. We evaluated growth and structures of 3D organoids treated with 5-fluorouracil ex vivo ., Results: Tumor-derived 3D organoids were grown successfully from 15 out of 21 patients (71.4%) and passaged with recapitulation of the histopathology of the original tumors. Successful formation of tumor-derived 3D organoids was associated significantly with poor response to presurgical neoadjuvant chemotherapy or chemoradiation therapy in informative patients ( P = 0.0357, progressive and stable diseases, n = 10 vs. partial response, n = 6). The 3D organoid formation capability and 5-fluorouracil resistance were accounted for by cancer cells with high CD44 expression and autophagy, respectively. Such cancer cells were found to be enriched in patient-derived 3D organoids surviving 5-fluorouracil treatment., Conclusions: The single cell-based 3D organoid system may serve as a highly efficient platform to explore cancer therapeutics and therapy resistance mechanisms in conjunction with morphological and functional assays with implications for translation in personalized medicine.
- Published
- 2018
- Full Text
- View/download PDF
14. Aggressive triple negative breast cancers have unique molecular signature on the basis of mitochondrial genetic and functional defects.
- Author
-
Guha M, Srinivasan S, Raman P, Jiang Y, Kaufman BA, Taylor D, Dong D, Chakrabarti R, Picard M, Carstens RP, Kijima Y, Feldman M, and Avadhani NG
- Subjects
- Cell Line, Tumor, Female, Humans, DNA, Mitochondrial genetics, DNA, Mitochondrial metabolism, Mitochondria genetics, Mitochondria metabolism, Mitochondria pathology, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology
- Abstract
Metastatic breast cancer is a leading cause of cancer-related deaths in women worldwide. Patients with triple negative breast cancer (TNBCs), a highly aggressive tumor subtype, have a particularly poor prognosis. Multiple reports demonstrate that altered content of the multicopy mitochondrial genome (mtDNA) in primary breast tumors correlates with poor prognosis. We earlier reported that mtDNA copy number reduction in breast cancer cell lines induces an epithelial-mesenchymal transition associated with metastasis. However, it is unknown whether the breast tumor subtypes (TNBC, Luminal and HER2+) differ in the nature and amount of mitochondrial defects and if mitochondrial defects can be used as a marker to identify tumors at risk for metastasis. By analyzing human primary tumors, cell lines and the TCGA dataset, we demonstrate a high degree of variability in mitochondrial defects among the tumor subtypes and TNBCs, in particular, exhibit higher frequency of mitochondrial defects, including reduced mtDNA content, mtDNA sequence imbalance (mtRNR1:ND4), impaired mitochondrial respiration and metabolic switch to glycolysis which is associated with tumorigenicity. We identified that genes involved in maintenance of mitochondrial structural and functional integrity are differentially expressed in TNBCs compared to non-TNBC tumors. Furthermore, we identified a subset of TNBC tumors that contain lower expression of epithelial splicing regulatory protein (ESRP)-1, typical of metastasizing cells. The overall impact of our findings reported here is that mitochondrial heterogeneity among TNBCs can be used to identify TNBC patients at risk of metastasis and the altered metabolism and metabolic genes can be targeted to improve chemotherapeutic response., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
15. Roles of Cytochrome P450 in Metabolism of Ethanol and Carcinogens.
- Author
-
Peter Guengerich F and Avadhani NG
- Subjects
- Animals, Endoplasmic Reticulum enzymology, Humans, Mice, Mitochondria, Liver enzymology, Oxidation-Reduction, Rats, Carcinogens metabolism, Cytochrome P-450 CYP2E1 metabolism, Ethanol metabolism
- Abstract
Cytochrome P450 (P450) enzymes are involved in the metabolism of carcinogens, as well as drugs, steroids, vitamins, and other classes of chemicals. P450s also oxidize ethanol, in particular P450 2E1. P450 2E1 oxidizes ethanol to acetaldehyde and then to acetic acid, roles also played by alcohol and aldehyde dehydrogenases. The role of P450 2E1 in cancer is complex in that P450 2E1 is also induced by ethanol, P450 2E1 is involved in the bioactivation and detoxication of a number of chemical carcinogens, and ethanol is an inhibitor of P450 2E1. Contrary to some literature, P450 2E1 expression and induction itself does not cause global oxidative stress in vivo, as demonstrated in studies using isoniazid treatment and gene deletion studies with rats and mice. However, a major fraction of P450 2E1 is localized in liver mitochondria instead of the endoplasmic reticulum, and studies with site-directed rat P450 2E1 mutants and natural human P450 2E1 N-terminal variants have shown that P450 2E1 localized in mitochondria is catalytically active and more proficient in producing reactive oxygen species and damage. The role of the mitochondrial oxidative stress in ethanol toxicity is still under investigation, as is the mechanism of altered electron transport to P450s that localize inside mitochondria instead of their typical endoplasmic reticulum environment.
- Published
- 2018
- Full Text
- View/download PDF
16. Autophagy supports generation of cells with high CD44 expression via modulation of oxidative stress and Parkin-mediated mitochondrial clearance.
- Author
-
Whelan KA, Chandramouleeswaran PM, Tanaka K, Natsuizaka M, Guha M, Srinivasan S, Darling DS, Kita Y, Natsugoe S, Winkler JD, Klein-Szanto AJ, Amaravadi RK, Avadhani NG, Rustgi AK, and Nakagawa H
- Subjects
- Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Epithelial-Mesenchymal Transition physiology, Esophageal Neoplasms metabolism, Esophageal Neoplasms pathology, Esophageal Squamous Cell Carcinoma, Humans, Keratinocytes metabolism, Keratinocytes physiology, Mitochondria metabolism, Oxidation-Reduction, RNA Interference physiology, Transforming Growth Factor beta metabolism, Autophagy physiology, Hyaluronan Receptors metabolism, Mitochondria physiology, Oxidative Stress physiology, Ubiquitin-Protein Ligases metabolism
- Abstract
High CD44 expression is associated with enhanced malignant potential in esophageal squamous cell carcinoma (ESCC), among the deadliest of all human carcinomas. Although alterations in autophagy and CD44 expression are associated with poor patient outcomes in various cancer types, the relationship between autophagy and cells with high CD44 expression remains incompletely understood. In transformed oesophageal keratinocytes, CD44
Low -CD24High (CD44L) cells give rise to CD44High -CD24-/Low (CD44H) cells via epithelial-mesenchymal transition (EMT) in response to transforming growth factor (TGF)-β. We couple patient samples and xenotransplantation studies with this tractable in vitro system of CD44L to CD44H cell conversion to investigate the functional role of autophagy in generation of cells with high CD44 expression. We report that high expression of the autophagy marker cleaved LC3 expression correlates with poor clinical outcome in ESCC. In ESCC xenograft tumours, pharmacological autophagy inhibition with chloroquine derivatives depletes cells with high CD44 expression while promoting oxidative stress. Autophagic flux impairment during EMT-mediated CD44L to CD44H cell conversion in vitro induces mitochondrial dysfunction, oxidative stress and cell death. During CD44H cell generation, transformed keratinocytes display evidence of mitophagy, including mitochondrial fragmentation, decreased mitochondrial content and mitochondrial translocation of Parkin, essential in mitophagy. RNA interference-mediated Parkin depletion attenuates CD44H cell generation. These data suggest that autophagy facilitates EMT-mediated CD44H generation via modulation of redox homeostasis and Parkin-dependent mitochondrial clearance. This is the first report to implicate mitophagy in regulation of tumour cells with high CD44 expression, representing a potential novel therapeutic avenue in cancers where EMT and CD44H cells have been implicated, including ESCC.- Published
- 2017
- Full Text
- View/download PDF
17. Mitochondrial dysfunction and mitochondrial dynamics-The cancer connection.
- Author
-
Srinivasan S, Guha M, Kashina A, and Avadhani NG
- Subjects
- Animals, Calcineurin physiology, Calcium Signaling, Cell Polarity, Cell Shape, Cytoskeleton metabolism, Cytoskeleton ultrastructure, DNA, Mitochondrial genetics, Humans, Membrane Potential, Mitochondrial, Mitochondria drug effects, Mitochondrial Dynamics drug effects, Mitochondrial Proteins physiology, Models, Biological, Neoplasm Proteins physiology, Neoplasms genetics, Quinazolinones pharmacology, Unfolded Protein Response, Cell Transformation, Neoplastic, Mitochondria metabolism, Mitochondrial Dynamics physiology, Neoplasms metabolism
- Abstract
Mitochondrial dysfunction is a hallmark of many diseases. The retrograde signaling initiated by dysfunctional mitochondria can bring about global changes in gene expression that alters cell morphology and function. Typically, this is attributed to disruption of important mitochondrial functions, such as ATP production, integration of metabolism, calcium homeostasis and regulation of apoptosis. Recent studies showed that in addition to these factors, mitochondrial dynamics might play an important role in stress signaling. Normal mitochondria are highly dynamic organelles whose size, shape and network are controlled by cell physiology. Defective mitochondrial dynamics play important roles in human diseases. Mitochondrial DNA defects and defective mitochondrial function have been reported in many cancers. Recent studies show that increased mitochondrial fission is a pro-tumorigenic phenotype. In this paper, we have explored the current understanding of the role of mitochondrial dynamics in pathologies. We present new data on mitochondrial dynamics and dysfunction to illustrate a causal link between mitochondrial DNA defects, excessive fission, mitochondrial retrograde signaling and cancer progression. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
18. Mitochondrial LON protease-dependent degradation of cytochrome c oxidase subunits under hypoxia and myocardial ischemia.
- Author
-
Sepuri NBV, Angireddy R, Srinivasan S, Guha M, Spear J, Lu B, Anandatheerthavarada HK, Suzuki CK, and Avadhani NG
- Subjects
- ATP-Dependent Proteases chemistry, ATP-Dependent Proteases genetics, Animals, Cyclic AMP-Dependent Protein Kinases metabolism, Humans, Male, Mice, Mitochondrial Proteins chemistry, Mitochondrial Proteins genetics, Models, Molecular, Phosphorylation, Protein Conformation, Protein Processing, Post-Translational, Protein Subunits, RAW 264.7 Cells, RNA Interference, RNA, Small Interfering genetics, Rabbits, Recombinant Proteins metabolism, ATP-Dependent Proteases metabolism, Cell Hypoxia physiology, Electron Transport Complex IV metabolism, Mitochondria, Heart enzymology, Mitochondrial Proteins metabolism, Myocardial Ischemia enzymology
- Abstract
The mitochondrial ATP dependent matrix protease, Lon, is involved in the maintenance of mitochondrial DNA nucleoids and degradation of abnormal or misfolded proteins. The Lon protease regulates mitochondrial Tfam (mitochondrial transcription factor A) level and thus modulates mitochondrial DNA (mtDNA) content. We have previously shown that hypoxic stress induces the PKA-dependent phosphorylation of cytochrome c oxidase (CcO) subunits I, IVi1, and Vb and a time-dependent reduction of these subunits in RAW 264.7 murine macrophages subjected to hypoxia and rabbit hearts subjected to ischemia/reperfusion. Here, we show that Lon is involved in the preferential turnover of phosphorylated CcO subunits under hypoxic/ischemic stress. Induction of Lon protease occurs at 6 to 12 h of hypoxia and this increase coincides with lower CcO subunit contents. Over-expression of flag-tagged wild type and phosphorylation site mutant Vb and IVi1 subunits (S40A and T52A, respectively) caused marked degradation of wild type protein under hypoxia while the mutant proteins were relatively resistant. Furthermore, the recombinant purified Lon protease degraded the phosphorylated IVi1 and Vb subunits, while the phosphorylation-site mutant proteins were resistant to degradation. 3D structural modeling shows that the phosphorylation sites are exposed to the matrix compartment, accessible to matrix PKA and Lon protease. Hypoxic stress did not alter CcO subunit levels in Lon depleted cells, confirming its role in CcO turnover. Our results therefore suggest that Lon preferentially degrades the phosphorylated subunits of CcO and plays a role in the regulation of CcO activity in hypoxia and ischemia/reperfusion injury., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
19. Blocking FSH induces thermogenic adipose tissue and reduces body fat.
- Author
-
Liu P, Ji Y, Yuen T, Rendina-Ruedy E, DeMambro VE, Dhawan S, Abu-Amer W, Izadmehr S, Zhou B, Shin AC, Latif R, Thangeswaran P, Gupta A, Li J, Shnayder V, Robinson ST, Yu YE, Zhang X, Yang F, Lu P, Zhou Y, Zhu LL, Oberlin DJ, Davies TF, Reagan MR, Brown A, Kumar TR, Epstein S, Iqbal J, Avadhani NG, New MI, Molina H, van Klinken JB, Guo EX, Buettner C, Haider S, Bian Z, Sun L, Rosen CJ, and Zaidi M
- Subjects
- Adipocytes drug effects, Adipocytes metabolism, Adipose Tissue drug effects, Adipose Tissue, Beige drug effects, Adipose Tissue, Beige metabolism, Adipose Tissue, White drug effects, Adipose Tissue, White metabolism, Animals, Antibodies immunology, Antibodies pharmacology, Diet, High-Fat adverse effects, Female, Follicle Stimulating Hormone, beta Subunit immunology, Haploinsufficiency, Male, Mice, Mitochondria drug effects, Mitochondria metabolism, Obesity drug therapy, Obesity prevention & control, Osteoporosis drug therapy, Ovariectomy, Oxygen Consumption drug effects, Receptors, FSH antagonists & inhibitors, Receptors, FSH genetics, Receptors, FSH metabolism, Uncoupling Protein 1 biosynthesis, Adipose Tissue metabolism, Adiposity drug effects, Follicle Stimulating Hormone, beta Subunit antagonists & inhibitors, Thermogenesis drug effects
- Abstract
Menopause is associated with bone loss and enhanced visceral adiposity. A polyclonal antibody that targets the β-subunit of the pituitary hormone follicle-stimulating hormone (Fsh) increases bone mass in mice. Here, we report that this antibody sharply reduces adipose tissue in wild-type mice, phenocopying genetic haploinsufficiency for the Fsh receptor gene Fshr. The antibody also causes profound beiging, increases cellular mitochondrial density, activates brown adipose tissue and enhances thermogenesis. These actions result from the specific binding of the antibody to the β-subunit of Fsh to block its action. Our studies uncover opportunities for simultaneously treating obesity and osteoporosis.
- Published
- 2017
- Full Text
- View/download PDF
20. Mitochondrial stress-induced p53 attenuates HIF-1α activity by physical association and enhanced ubiquitination.
- Author
-
Chowdhury AR, Long A, Fuchs SY, Rustgi A, and Avadhani NG
- Subjects
- A549 Cells, Animals, Base Sequence, COS Cells, Cell Line, Tumor, HCT116 Cells, Humans, Mice, Protein Binding, Rats, Sequence Deletion, Signal Transduction, Ubiquitination, Calcineurin metabolism, Calcium metabolism, DNA, Mitochondrial genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Mitochondria genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Retrograde signaling is a mechanism by which mitochondrial dysfunction is communicated to the nucleus for inducing a metabolic shift essential for cell survival. Previously, we showed that partial mitochondrial DNA (mtDNA) depletion in different cell types induced mitochondrial retrograde signaling pathway (MtRS) involving Ca
+2 -sensitive Calcineurin (Cn) activation as an immediate upstream event of stress response. In multiple cell types, this stress signaling was shown to induce tumorigenic phenotypes in immortalized cells. In this study we show that MtRS also induces p53 expression, which was abrogated by Ca2+ chelators and short hairpin RNA-mediated knockdown of CnAβ mRNA. Mitochondrial dysfunction induced by mitochondrial ionophore, carbonyl cyanide m-chlorophenyl hydrazone and other respiratory inhibitors, which perturb the transmembrane potential, were equally efficient in inducing the expression of p53 and downregulation of MDM2. Stress-induced p53 physically interacted with hypoxia-inducible factor-1α (HIF-1α) and attenuated the latter's binding to promoter DNA motifs. In addition, p53 promoted ubiquitination and degradation of HIF-1α in partial mtDNA-depleted cells. The mtDNA depleted cells, with inhibited HIF-1α, showed upregulation of glycolytic pathway genes, glucose transporter 1-4 (Glut1-4), phosphoglycerate kinase 1 and Glucokinase but not of prolyl hydroxylase isoforms. For the first time we show that p53 is induced as part of MtRS and it renders HIF-1α inactive by physical interaction. In this respect, our results show that MtRS induces tumor growth independent of the HIF-1α pathway., Competing Interests: The authors declare no conflict of the interest.- Published
- 2017
- Full Text
- View/download PDF
21. β -Naphthoflavone-Induced Mitochondrial Respiratory Damage in Cyp1 Knockout Mouse and in Cell Culture Systems: Attenuation by Resveratrol Treatment.
- Author
-
Anandasadagopan SK, Singh NM, Raza H, Bansal S, Selvaraj V, Singh S, Chowdhury AR, Leu NA, and Avadhani NG
- Subjects
- Animals, Cell Culture Techniques, Male, Mice, Mice, Knockout, Resveratrol, Stilbenes pharmacology, Cytochrome P-450 CYP1A1 genetics, Mitochondria metabolism, Stilbenes therapeutic use, beta-Naphthoflavone metabolism
- Abstract
A number of xenobiotic-inducible cytochrome P450s (CYPs) are now known to be localized in the mitochondrial compartment, though their pharmacological or toxicological roles remain unclear. Here, we show that BNF treatment markedly inhibits liver mitochondrial O
2 consumption rate (OCR), ADP-dependent OCR, and also reserve OCR, in wild-type mice but not in Cyp1a1/1a2(-/-) double knockout mice. BNF treatment markedly affected mitochondrial complex I and complex IV activities and also attenuated mitochondrial gene expression. Furthermore, under in vitro conditions, BNF treatment induced cellular ROS production, which was inhibited by mitochondria-targeted antioxidant Mito-CP and CYP inhibitor proadefin, suggesting that most of the ROS production was intramitochondrial and probably involved the catalytic activity of mitochondrial CYP1 enzymes. Interestingly, our results also show that the AHR antagonist resveratrol, markedly attenuated BNF-induced liver mitochondrial defects in wild-type mice, confirming the role of AHR and AHR-regulated CYP1 genes in eliciting mitochondrial dysfunction. These results are consistent with reduced BNF-induced mitochondrial toxicity in Cyp1a1/1a2(-/-) mice and elevated ROS production in COS cells stably expressing CYP1A1. We propose that increased mitochondrial ROS production and respiratory dysfunction are part of xenobiotic toxicity. Resveratrol, a chemopreventive agent, renders protection against BNF-induced toxicity.- Published
- 2017
- Full Text
- View/download PDF
22. HnRNPA2 is a novel histone acetyltransferase that mediates mitochondrial stress-induced nuclear gene expression.
- Author
-
Guha M, Srinivasan S, Guja K, Mejia E, Garcia-Diaz M, Johnson FB, Ruthel G, Kaufman BA, Rappaport EF, Glineburg MR, Fang JK, Klein-Szanto AJ, Klein Szanto A, Nakagawa H, Basha J, Kundu T, and Avadhani NG
- Abstract
Reduced mitochondrial DNA copy number, mitochondrial DNA mutations or disruption of electron transfer chain complexes induce mitochondria-to-nucleus retrograde signaling, which induces global change in nuclear gene expression ultimately contributing to various human pathologies including cancer. Recent studies suggest that these mitochondrial changes cause transcriptional reprogramming of nuclear genes although the mechanism of this cross talk remains unclear. Here, we provide evidence that mitochondria-to-nucleus retrograde signaling regulates chromatin acetylation and alters nuclear gene expression through the heterogeneous ribonucleoprotein A2 (hnRNAP2). These processes are reversed when mitochondrial DNA content is restored to near normal cell levels. We show that the mitochondrial stress-induced transcription coactivator hnRNAP2 acetylates Lys 8 of H4 through an intrinsic histone lysine acetyltransferase (KAT) activity with Arg 48 and Arg 50 of hnRNAP2 being essential for acetyl-CoA binding and acetyltransferase activity. H4K8 acetylation at the mitochondrial stress-responsive promoters by hnRNAP2 is essential for transcriptional activation. We found that the previously described mitochondria-to-nucleus retrograde signaling-mediated transformation of C2C12 cells caused an increased expression of genes involved in various oncogenic processes, which is retarded in hnRNAP2 silenced or hnRNAP2 KAT mutant cells. Taken together, these data show that altered gene expression by mitochondria-to-nucleus retrograde signaling involves a novel hnRNAP2-dependent epigenetic mechanism that may have a role in cancer and other pathologies.
- Published
- 2016
- Full Text
- View/download PDF
23. Targeting mitochondrial biogenesis to overcome drug resistance to MAPK inhibitors.
- Author
-
Zhang G, Frederick DT, Wu L, Wei Z, Krepler C, Srinivasan S, Chae YC, Xu X, Choi H, Dimwamwa E, Ope O, Shannan B, Basu D, Zhang D, Guha M, Xiao M, Randell S, Sproesser K, Xu W, Liu J, Karakousis GC, Schuchter LM, Gangadhar TC, Amaravadi RK, Gu M, Xu C, Ghosh A, Xu W, Tian T, Zhang J, Zha S, Liu Q, Brafford P, Weeraratna A, Davies MA, Wargo JA, Avadhani NG, Lu Y, Mills GB, Altieri DC, Flaherty KT, and Herlyn M
- Subjects
- Animals, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Extracellular Signal-Regulated MAP Kinases genetics, Extracellular Signal-Regulated MAP Kinases metabolism, Female, HSP90 Heat-Shock Proteins antagonists & inhibitors, HSP90 Heat-Shock Proteins genetics, HSP90 Heat-Shock Proteins metabolism, Humans, Male, Melanoma genetics, Melanoma metabolism, Melanoma pathology, Mice, Mitochondria genetics, Mitochondria pathology, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Transcription Factors genetics, Transcription Factors metabolism, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Guanidines pharmacology, Lactams, Macrocyclic pharmacology, Melanoma drug therapy, Mitochondria metabolism, Mitochondrial Dynamics drug effects, Neoplasm Proteins antagonists & inhibitors, Protein Kinase Inhibitors pharmacology
- Abstract
Targeting multiple components of the MAPK pathway can prolong the survival of patients with BRAFV600E melanoma. This approach is not curative, as some BRAF-mutated melanoma cells are intrinsically resistant to MAPK inhibitors (MAPKi). At the systemic level, our knowledge of how signaling pathways underlie drug resistance needs to be further expanded. Here, we have shown that intrinsically resistant BRAF-mutated melanoma cells with a low basal level of mitochondrial biogenesis depend on this process to survive MAPKi. Intrinsically resistant cells exploited an integrated stress response, exhibited an increase in mitochondrial DNA content, and required oxidative phosphorylation to meet their bioenergetic needs. We determined that intrinsically resistant cells rely on the genes encoding TFAM, which controls mitochondrial genome replication and transcription, and TRAP1, which regulates mitochondrial protein folding. Therefore, we targeted mitochondrial biogenesis with a mitochondrium-targeted, small-molecule HSP90 inhibitor (Gamitrinib), which eradicated intrinsically resistant cells and augmented the efficacy of MAPKi by inducing mitochondrial dysfunction and inhibiting tumor bioenergetics. A subset of tumor biopsies from patients with disease progression despite MAPKi treatment showed increased mitochondrial biogenesis and tumor bioenergetics. A subset of acquired drug-resistant melanoma cell lines was sensitive to Gamitrinib. Our study establishes mitochondrial biogenesis, coupled with aberrant tumor bioenergetics, as a potential therapy escape mechanism and paves the way for a rationale-based combinatorial strategy to improve the efficacy of MAPKi.
- Published
- 2016
- Full Text
- View/download PDF
24. Disruption of cytochrome c oxidase function induces the Warburg effect and metabolic reprogramming.
- Author
-
Srinivasan S, Guha M, Dong DW, Whelan KA, Ruthel G, Uchikado Y, Natsugoe S, Nakagawa H, and Avadhani NG
- Subjects
- Animals, Cell Line, Electron Transport Complex IV genetics, Gene Silencing, Mice, Oxidative Stress, Signal Transduction, Electron Transport Complex IV metabolism
- Abstract
Defects in mitochondrial oxidative phosphorylation complexes, altered bioenergetics and metabolic shift are often seen in cancers. Here we show a role for the dysfunction of the electron transport chain component cytochrome c oxidase (CcO) in cancer progression. We show that genetic silencing of the CcO complex by shRNA expression and loss of CcO activity in multiple cell types from the mouse and human sources resulted in metabolic shift to glycolysis, loss of anchorage-dependent growth and acquired invasive phenotypes. Disruption of the CcO complex caused loss of transmembrane potential and induction of Ca2+/Calcineurin-mediated retrograde signaling. Propagation of this signaling includes activation of PI3-kinase, IGF1R and Akt, Ca2(+)-sensitive transcription factors and also TGFβ1, MMP16 and periostin, which are involved in oncogenic progression. Whole-genome expression analysis showed the upregulation of genes involved in cell signaling, extracellular matrix interactions, cell morphogenesis, cell motility and migration. The transcription profiles reveal extensive similarity to retrograde signaling initiated by partial mitochondrial DNA depletion, although distinct differences are observed in signaling induced by CcO dysfunction. The possible CcO dysfunction as a biomarker for cancer progression was supported by data showing that esophageal tumors from human patients show reduced CcO subunits IVi1 and Vb in regions that were previously shown to be the hypoxic core of the tumors. Our results show that mitochondrial electron transport chain defect initiates a retrograde signaling. These results suggest that a defect in the CcO complex can potentially induce tumor progression.
- Published
- 2016
- Full Text
- View/download PDF
25. ALDH2 modulates autophagy flux to regulate acetaldehyde-mediated toxicity thresholds.
- Author
-
Tanaka K, Whelan KA, Chandramouleeswaran PM, Kagawa S, Rustgi SL, Noguchi C, Guha M, Srinivasan S, Amanuma Y, Ohashi S, Muto M, Klein-Szanto AJ, Noguchi E, Avadhani NG, and Nakagawa H
- Abstract
A polymorphic mutation in the acetaldehyde dehydrogenase 2 (ALDH2) gene has been epidemiologically linked to the high susceptibility to esophageal carcinogenesis for individuals with alcohol use disorders. Mice subjected to alcohol drinking show increased oxidative stress and DNA adduct formation in esophageal epithelia where Aldh2 loss augments alcohol-induced genotoxic effects; however, it remains elusive as to how esophageal epithelial cells with dysfunctional Aldh2 cope with oxidative stress related to alcohol metabolism. Here, we investigated the role of autophagy in murine esophageal epithelial cells (keratinocytes) exposed to ethanol and acetaldehyde. We find that ethanol and acetaldehyde trigger oxidative stress via mitochondrial superoxide in esophageal keratinocytes. Aldh2-deficient cells appeared to be highly susceptible to ethanol- or acetaldehyde-mediated toxicity. Alcohol dehydrogenase-mediated acetaldehyde production was implicated in ethanol-induced cell injury in Aldh2 deficient cells as ethanol-induced oxidative stress and cell death was partially inhibited by 4-methylpyrazole. Acetaldehyde activated autophagy flux in esophageal keratinocytes where Aldh2 deficiency increased dependence on autophagy to cope with ethanol-induced acetaldehyde-mediated oxidative stress. Pharmacological inhibition of autophagy flux by chloroquine stabilized p62/SQSTM1, and increased basal and acetaldehyde-mediate oxidative stress in Aldh2 deficient cells as documented in monolayer culture as well as single-cell derived three-dimensional esophageal organoids, recapitulating a physiological esophageal epithelial proliferation-differentiation gradient. Our innovative approach indicates, for the first time, that autophagy may provide cytoprotection to esophageal epithelial cells responding to oxidative stress that is induced by ethanol and its major metabolite acetaldehyde. Defining autophagymediated cytoprotection against alcohol-induced genotoxicity in the context of Aldh2 deficiency, our study provides mechanistic insights into the tumor suppressor functions of ALDH2 and autophagy in alcohol-related esophageal carcinogenesis.
- Published
- 2016
26. Enhanced osteoclastogenesis by mitochondrial retrograde signaling through transcriptional activation of the cathepsin K gene.
- Author
-
Guha M, Srinivasan S, Koenigstein A, Zaidi M, and Avadhani NG
- Subjects
- Animals, CCAAT-Enhancer-Binding Protein-delta antagonists & inhibitors, CCAAT-Enhancer-Binding Protein-delta genetics, CCAAT-Enhancer-Binding Protein-delta metabolism, Cathepsin K antagonists & inhibitors, Cathepsin K chemistry, Cathepsin K genetics, Cell Hypoxia, Cyclic AMP Response Element-Binding Protein antagonists & inhibitors, Cyclic AMP Response Element-Binding Protein genetics, Cyclic AMP Response Element-Binding Protein metabolism, Genes, Reporter, Heterogeneous-Nuclear Ribonucleoprotein Group A-B antagonists & inhibitors, Heterogeneous-Nuclear Ribonucleoprotein Group A-B genetics, Heterogeneous-Nuclear Ribonucleoprotein Group A-B metabolism, Mice, Mitochondria enzymology, NFATC Transcription Factors antagonists & inhibitors, NFATC Transcription Factors genetics, NFATC Transcription Factors metabolism, Osteoclasts cytology, Osteoclasts enzymology, RANK Ligand metabolism, RAW 264.7 Cells, RNA Interference, RNA, Messenger metabolism, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Transcriptional Activation, Cathepsin K metabolism, Mitochondria metabolism, Osteoclasts metabolism, Osteogenesis, Promoter Regions, Genetic, Signal Transduction
- Abstract
Mitochondrial dysfunction has emerged as an important factor in wide ranging human pathologies. We have previously defined a retrograde signaling pathway that originates from dysfunctional mitochondria (Mt-RS) and causes a global nuclear transcriptional reprograming as its end point. Mitochondrial dysfunction causing disruption of mitochondrial membrane potential and consequent increase in cytosolic calcium [Ca(2) ](c) activates calcineurin and the transcription factors NF-κB, NFAT, CREB, and C/EBPδ. In macrophages, this signaling complements receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastic differentiation. Here, we show that the Mt-RS activated transcriptional coactivator heterogeneous ribonucleoprotein A2 (hnRNP A2) is induced by hypoxia in murine macrophages. We demonstrate that the cathepsin K gene (Ctsk), one of the key genes upregulated during osteoclast differentiation, is transcriptionally activated by Mt-RS factors. HnRNP A2 acts as a coactivator with nuclear transcription factors, cRel, and C/EBPδ for Ctsk promoter activation under hypoxic conditions. Notably, our study shows that hypoxia-induced activation of the stress target factors mediates effects similar to that of RANKL with regard to Ctsk activation. We therefore suggest that mitochondrial dysfunction and activation of Mt-RS, induced by various pathophysiologic conditions, is a potential risk factor for osteoclastogenesis and bone loss., (© 2015 New York Academy of Sciences.)
- Published
- 2016
- Full Text
- View/download PDF
27. Mitochondrial respiratory defects promote the Warburg effect and cancer progression.
- Author
-
Srinivasan S, Guha M, and Avadhani NG
- Abstract
In the past decade mitochondria have emerged as an important cellular signaling hub controlling metabolism, epigenetics, and cell fate. Dysfunctional mitochondria initiate a retrograde nuclear response that influences the cellular reprograming observed in various human pathologies, including cancer. New data suggest that loss of cytochrome c oxidase function promotes the Warburg effect and upregulates several genes with roles in tumor development.
- Published
- 2015
- Full Text
- View/download PDF
28. Mitochondrial SOD2 regulates epithelial-mesenchymal transition and cell populations defined by differential CD44 expression.
- Author
-
Kinugasa H, Whelan KA, Tanaka K, Natsuizaka M, Long A, Guo A, Chang S, Kagawa S, Srinivasan S, Guha M, Yamamoto K, St Clair DK, Avadhani NG, Diehl JA, and Nakagawa H
- Subjects
- Cell Line, Gene Expression Regulation, Enzymologic, Homeodomain Proteins metabolism, Humans, Hyaluronan Receptors, Mitochondria enzymology, NF-kappa B metabolism, Repressor Proteins metabolism, Zinc Finger E-box Binding Homeobox 2, Epithelial-Mesenchymal Transition, Superoxide Dismutase physiology
- Abstract
Epithelial-mesenchymal transition (EMT) promotes cancer cell invasion, metastasis and treatment failure. EMT may be activated in cancer cells by reactive oxygen species (ROS). EMT may promote conversion of a subset of cancer cells from a CD44(low)-CD24(high) (CD44L) epithelial phenotype to a CD44(high)-CD24(-/low) (CD44H) mesenchymal phenotype, the latter associated with increased malignant properties of cancer cells. ROS are required for cells undergoing EMT, although excessive ROS may induce cell death or senescence; however, little is known as to how cellular antioxidant capabilities may be regulated during EMT. Mitochondrial superoxide dismutase 2 (SOD2) is frequently overexpressed in oral and esophageal cancers. Here, we investigate mechanisms of SOD2 transcriptional regulation in EMT, as well as the functional role of this antioxidant in EMT. Using well-characterized genetically engineered oral and esophageal human epithelial cell lines coupled with RNA interference and flow cytometric approaches, we find that transforming growth factor (TGF)-β stimulates EMT, resulting in conversion of CD44L to CD44H cells, the latter of which display SOD2 upregulation. SOD2 induction in transformed keratinocytes was concurrent with suppression of TGF-β-mediated induction of both ROS and senescence. SOD2 gene expression appeared to be transcriptionally regulated by NF-κB and ZEB2, but not ZEB1. Moreover, SOD2-mediated antioxidant activity may restrict conversion of CD44L cells to CD44H cells at the early stages of EMT. These data provide novel mechanistic insights into the dynamic expression of SOD2 during EMT. In addition, we delineate a functional role for SOD2 in EMT via the influence of this antioxidant upon distinct CD44L and CD44H subsets of cancer cells that have been implicated in oral and esophageal tumor biology.
- Published
- 2015
- Full Text
- View/download PDF
29. Defects in cytochrome c oxidase expression induce a metabolic shift to glycolysis and carcinogenesis.
- Author
-
Dong DW, Srinivasan S, Guha M, and Avadhani NG
- Abstract
Mitochondrial metabolic dysfunction is often seen in cancers. This paper shows that the defect in a mitochondrial electron transport component, the cytochrome c oxidase (CcO), leads to increased glycolysis and carcinogenesis. Using whole genome microarray expression analysis we show that genetic silencing of the CcO subunit Cox4i1 in mouse C2C12 myoblasts resulted in metabolic shift to glycolysis, activated a retrograde stress signaling, and induced carcinogenesis. In the knockdown cells, the expression of Cox4i1 was less than 5% of the control and the expression of the irreversible glycolytic enzymes (Hk1, Pfkm and Pkm) increased two folds, facilitating metabolic shift to glycolysis. The expression of Ca (2+) sensitive Calcineurin (Ppp3ca) and the expression of PI3-kinase (Pik3r4 and Pik3cb) increased by two folds. This Ca (2+)/Calcineurin/PI3K retrograde stress signaling induced the up-regulation of many nuclear genes involved in tumor progression. Overall, we found 1047 genes with 2-folds expression change (with p-value less than 0.01) between the knockdown and the control, among which were 35 up-regulated genes in pathways in cancer (enrichment p-value less than 10(- 5)). Functional analysis revealed that the up-regulated genes in pathways in cancer were dominated by genes in signal transduction, regulation of transcription and PI3K signaling pathway. These results suggest that a defect in CcO complex initiates a retrograde signaling which can induce tumor progression. Physiological studies of these cells and esophageal tumors from human patients support these results. GEO accession number = GSE68525.
- Published
- 2015
- Full Text
- View/download PDF
30. Mitochondrial retrograde signaling induces epithelial-mesenchymal transition and generates breast cancer stem cells.
- Author
-
Guha M, Srinivasan S, Ruthel G, Kashina AK, Carstens RP, Mendoza A, Khanna C, Van Winkle T, and Avadhani NG
- Subjects
- Animals, Breast Neoplasms metabolism, Breast Neoplasms pathology, Calcineurin genetics, Calcineurin metabolism, Cell Line, Cell Movement genetics, DNA, Mitochondrial metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Epithelial Cells metabolism, Female, Gene Dosage, Gene Expression, Humans, Immunoblotting, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms secondary, MCF-7 Cells, Mice, SCID, Microscopy, Confocal, Mitochondria genetics, Mitochondria metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Neoplastic Stem Cells pathology, Oxygen Consumption genetics, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Transcription Factors genetics, Transcription Factors metabolism, Transplantation, Heterologous, Breast Neoplasms genetics, DNA, Mitochondrial genetics, Epithelial-Mesenchymal Transition genetics, Neoplastic Stem Cells metabolism, Signal Transduction genetics
- Abstract
Metastatic breast tumors undergo epithelial-to-mesenchymal transition (EMT), which renders them resistant to therapies targeted to the primary cancers. The mechanistic link between mtDNA (mitochondrial DNA) reduction, often seen in breast cancer patients, and EMT is unknown. We demonstrate that reducing mtDNA content in human mammary epithelial cells (hMECs) activates Calcineurin (Cn)-dependent mitochondrial retrograde signaling pathway, which induces EMT-like reprogramming to fibroblastic morphology, loss of cell polarity, contact inhibition and acquired migratory and invasive phenotype. Notably, mtDNA reduction generates breast cancer stem cells. In addition to retrograde signaling markers, there is an induction of mesenchymal genes but loss of epithelial markers in these cells. The changes are reversed by either restoring the mtDNA content or knockdown of CnAα mRNA, indicating the causal role of retrograde signaling in EMT. Our results point to a new therapeutic strategy for metastatic breast cancers targeted to the mitochondrial retrograde signaling pathway for abrogating EMT and attenuating cancer stem cells, which evade conventional therapies. We report a novel regulatory mechanism by which low mtDNA content generates EMT and cancer stem cells in hMECs.
- Published
- 2014
- Full Text
- View/download PDF
31. Targeting of splice variants of human cytochrome P450 2C8 (CYP2C8) to mitochondria and their role in arachidonic acid metabolism and respiratory dysfunction.
- Author
-
Bajpai P, Srinivasan S, Ghosh J, Nagy LD, Wei S, Guengerich FP, and Avadhani NG
- Subjects
- Amino Acid Sequence, Amino Acids metabolism, Animals, Arachidonic Acid metabolism, Aryl Hydrocarbon Hydroxylases chemistry, Aryl Hydrocarbon Hydroxylases metabolism, Biocatalysis, COS Cells, Cell Respiration, Chlorocebus aethiops, Computer Simulation, Cytochrome P-450 CYP2C8 chemistry, Cytochrome P-450 CYP2C8 metabolism, Heme metabolism, Hep G2 Cells, Humans, Isoenzymes metabolism, Microsomes, Liver enzymology, Models, Molecular, Molecular Sequence Data, Oxidation-Reduction, Oxidative Stress, Protein Binding, Protein Transport, RNA, Messenger genetics, RNA, Messenger metabolism, Reactive Oxygen Species metabolism, Sequence Alignment, Alternative Splicing genetics, Aryl Hydrocarbon Hydroxylases genetics, Cytochrome P-450 CYP2C8 genetics, Mitochondria metabolism, Mitochondria pathology
- Abstract
In this study, we found that the full-length CYP2C8 (WT CYP2C8) and N-terminal truncated splice variant 3 (∼ 44-kDa mass) are localized in mitochondria in addition to the endoplasmic reticulum. Analysis of human livers showed that the mitochondrial levels of these two forms varied markedly. Molecular modeling based on the x-ray crystal structure coordinates of CYP2D6 and CYP2C8 showed that despite lacking the N-terminal 102 residues variant 3 possessed nearly complete substrate binding and heme binding pockets. Stable expression of cDNAs in HepG2 cells showed that the WT protein is mostly targeted to the endoplasmic reticulum and at low levels to mitochondria, whereas variant 3 is primarily targeted to mitochondria and at low levels to the endoplasmic reticulum. Enzyme reconstitution experiments showed that both microsomal and mitochondrial WT CYP2C8 efficiently catalyzed paclitaxel 6-hydroxylation. However, mitochondrial variant 3 was unable to catalyze this reaction possibly because of its inability to stabilize the large 854-Da substrate. Conversely, mitochondrial variant 3 catalyzed the metabolism of arachidonic acid into 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid when reconstituted with adrenodoxin and adrenodoxin reductase. HepG2 cells stably expressing variant 3 generated higher levels of reactive oxygen species and showed a higher level of mitochondrial respiratory dysfunction. This study suggests that mitochondrially targeted variant 3 CYP2C8 may contribute to oxidative stress in various tissues., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
32. Mitochondrial targeting of cytochrome P450 (CYP) 1B1 and its role in polycyclic aromatic hydrocarbon-induced mitochondrial dysfunction.
- Author
-
Bansal S, Leu AN, Gonzalez FJ, Guengerich FP, Chowdhury AR, Anandatheerthavarada HK, and Avadhani NG
- Subjects
- Adrenodoxin genetics, Adrenodoxin metabolism, Animals, Aryl Hydrocarbon Hydroxylases genetics, Benzo(a)pyrene pharmacology, COS Cells, Cell Line, Tumor, Chlorocebus aethiops, Cytochrome P-450 CYP1B1, Female, Humans, Male, Mice, Mice, Knockout, Mitochondria genetics, Mitochondria pathology, Mutagenesis, Oxidation-Reduction drug effects, Polychlorinated Dibenzodioxins pharmacology, Protein Transport drug effects, Aryl Hydrocarbon Hydroxylases metabolism, Benzo(a)pyrene adverse effects, Mitochondria enzymology, Oxygen Consumption drug effects, Polychlorinated Dibenzodioxins adverse effects, Protein Sorting Signals, Teratogens
- Abstract
We report that polycyclic aromatic hydrocarbon (PAH)-inducible CYP1B1 is targeted to mitochondria by sequence-specific cleavage at the N terminus by a cytosolic Ser protease (polyserase 1) to activate the cryptic internal signal. Site-directed mutagenesis, COS-7 cell transfection, and in vitro import studies in isolated mitochondria showed that a positively charged domain at residues 41-48 of human CYP1B1 is part of the mitochondrial (mt) import signal. Ala scanning mutations showed that the Ser protease cleavage site resides between residues 37 and 41 of human CYP1B1. Benzo[a]pyrene (BaP) treatment induced oxidative stress, mitochondrial respiratory defects, and mtDNA damage that was attenuated by a CYP1B1-specific inhibitor, 2,3,4,5-tetramethoxystilbene. In support, the mitochondrial CYP1B1 supported by mitochondrial ferredoxin (adrenodoxin) and ferredoxin reductase showed high aryl hydrocarbon hydroxylase activity. Administration of benzo[a]pyrene or 2,3,7,8-tetrachlorodibenzodioxin induced similar mitochondrial functional abnormalities and oxidative stress in the lungs of wild-type mice and Cyp1a1/1a2-null mice, but the effects were markedly blunted in Cyp1b1-null mice. These results confirm a role for CYP1B1 in inducing PAH-mediated mitochondrial dysfunction. The role of mitochondrial CYP1B1 was assessed using A549 lung epithelial cells stably expressing shRNA against NADPH-cytochrome P450 oxidoreductase or mitochondrial adrenodoxin. Our results not only show conservation of the endoprotease cleavage mechanism for mitochondrial import of family 1 CYPs but also reveal a direct role for mitochondrial CYP1B1 in PAH-mediated oxidative and chemical damage to mitochondria.
- Published
- 2014
- Full Text
- View/download PDF
33. Triggering ubiquitination of IFNAR1 protects tissues from inflammatory injury.
- Author
-
Bhattacharya S, Katlinski KV, Reichert M, Takano S, Brice A, Zhao B, Yu Q, Zheng H, Carbone CJ, Katlinskaya YV, Leu NA, McCorkell KA, Srinivasan S, Girondo M, Rui H, May MJ, Avadhani NG, Rustgi AK, and Fuchs SY
- Subjects
- Acute Disease, Animals, Bone Marrow Transplantation, Chemical and Drug Induced Liver Injury pathology, Chemical and Drug Induced Liver Injury surgery, Chemical and Drug Induced Liver Injury veterinary, Chronic Disease, Female, Gene Knock-In Techniques, Interleukin-1beta metabolism, Interleukin-6 metabolism, Liver physiology, Mice, Mice, Inbred C57BL, Pancreas physiology, Pancreatitis chemically induced, Pancreatitis pathology, Pancreatitis surgery, Receptor, Interferon alpha-beta genetics, Regeneration, Tumor Necrosis Factor-alpha metabolism, Ubiquitination, Receptor, Interferon alpha-beta metabolism
- Abstract
Type 1 interferons (IFN) protect the host against viruses by engaging a cognate receptor (consisting of IFNAR1/IFNAR2 chains) and inducing downstream signaling and gene expression. However, inflammatory stimuli can trigger IFNAR1 ubiquitination and downregulation thereby attenuating IFN effects in vitro. The significance of this paradoxical regulation is unknown. Presented here results demonstrate that inability to stimulate IFNAR1 ubiquitination in the Ifnar1(SA) knock-in mice renders them highly susceptible to numerous inflammatory syndromes including acute and chronic pancreatitis, and autoimmune and toxic hepatitis. Ifnar1(SA) mice (or their bone marrow-receiving wild type animals) display persistent immune infiltration of inflamed tissues, extensive damage and gravely inadequate tissue regeneration. Pharmacologic stimulation of IFNAR1 ubiquitination is protective against from toxic hepatitis and fulminant generalized inflammation in wild type but not Ifnar1(SA) mice. These results suggest that endogenous mechanisms that trigger IFNAR1 ubiquitination for limiting the inflammation-induced tissue damage can be purposely mimicked for therapeutic benefits.
- Published
- 2014
- Full Text
- View/download PDF
34. Mitochondrial retrograde signaling at the crossroads of tumor bioenergetics, genetics and epigenetics.
- Author
-
Guha M and Avadhani NG
- Subjects
- Humans, Neoplasms genetics, Energy Metabolism, Epigenesis, Genetic, Mitochondria metabolism, Neoplasms metabolism, Signal Transduction
- Abstract
Mitochondria play a central role not only in energy production but also in the integration of metabolic pathways as well as signals for apoptosis and autophagy. It is becoming increasingly apparent that mitochondria in mammalian cells play critical roles in the initiation and propagation of various signaling cascades. In particular, mitochondrial metabolic and respiratory states and status on mitochondrial genetic instability are communicated to the nucleus as an adaptive response through retrograde signaling. Each mammalian cell contains multiple copies of the mitochondrial genome (mtDNA). A reduction in mtDNA copy number has been reported in various human pathological conditions such as diabetes, obesity, neurodegenerative disorders, aging and cancer. Reduction in mtDNA copy number disrupts mitochondrial membrane potential (Δψm) resulting in dysfunctional mitochondria. Dysfunctional mitochondria trigger retrograde signaling and communicate their changing metabolic and functional state to the nucleus as an adaptive response resulting in an altered nuclear gene expression profile and altered cell physiology and morphology. In this review, we provide an overview of the various modes of mitochondrial retrograde signaling focusing particularly on the Ca(2+)/Calcineurin mediated retrograde signaling. We discuss the contribution of the key factors of the pathway such as Calcineurin, IGF1 receptor, Akt kinase and HnRNPA2 in the propagation of signaling and their role in modulating genetic and epigenetic changes favoring cellular reprogramming towards tumorigenesis., (© 2013. Published by Elsevier B.V. and Mitochondria Research Society.)
- Published
- 2013
- Full Text
- View/download PDF
35. Oxidative stress induced mitochondrial protein kinase A mediates cytochrome c oxidase dysfunction.
- Author
-
Srinivasan S, Spear J, Chandran K, Joseph J, Kalyanaraman B, and Avadhani NG
- Subjects
- Animals, Antioxidants pharmacology, Cell Hypoxia drug effects, Cell Line, Cell Respiration drug effects, Electron Transport Complex IV genetics, Enzyme Activation drug effects, Mice, Mitochondria drug effects, Mitochondria metabolism, Mutation, Myocardial Ischemia enzymology, Myocardial Ischemia metabolism, Myocardial Ischemia pathology, Phosphorylation drug effects, Protein Subunits genetics, Protein Subunits metabolism, Protein Transport drug effects, Proteolysis drug effects, Reactive Oxygen Species metabolism, Reperfusion Injury enzymology, Reperfusion Injury metabolism, Reperfusion Injury pathology, Cyclic AMP-Dependent Protein Kinases metabolism, Electron Transport Complex IV metabolism, Mitochondria enzymology, Oxidative Stress drug effects
- Abstract
Previously we showed that Protein kinase A (PKA) activated in hypoxia and myocardial ischemia/reperfusion mediates phosphorylation of subunits I, IVi1 and Vb of cytochrome c oxidase. However, the mechanism of activation of the kinase under hypoxia remains unclear. It is also unclear if hypoxic stress activated PKA is different from the cAMP dependent mitochondrial PKA activity reported under normal physiological conditions. In this study using RAW 264.7 macrophages and in vitro perfused mouse heart system we investigated the nature of PKA activated under hypoxia. Limited protease treatment and digitonin fractionation of intact mitochondria suggests that higher mitochondrial PKA activity under hypoxia is mainly due to increased sequestration of PKA Catalytic α (PKAα) subunit in the mitochondrial matrix compartment. The increase in PKA activity is independent of mitochondrial cAMP and is not inhibited by adenylate cyclase inhibitor, KH7. Instead, activation of hypoxia-induced PKA is dependent on reactive oxygen species (ROS). H89, an inhibitor of PKA activity and the antioxidant Mito-CP prevented loss of CcO activity in macrophages under hypoxia and in mouse heart under ischemia/reperfusion injury. Substitution of wild type subunit Vb of CcO with phosphorylation resistant S40A mutant subunit attenuated the loss of CcO activity and reduced ROS production. These results provide a compelling evidence for hypoxia induced phosphorylation as a signal for CcO dysfunction. The results also describe a novel mechanism of mitochondrial PKA activation which is independent of mitochondrial cAMP, but responsive to ROS.
- Published
- 2013
- Full Text
- View/download PDF
36. Mitochondria-targeted heme oxygenase-1 induces oxidative stress and mitochondrial dysfunction in macrophages, kidney fibroblasts and in chronic alcohol hepatotoxicity.
- Author
-
Bansal S, Biswas G, and Avadhani NG
- Subjects
- Animals, COS Cells, Cell Hypoxia, Chlorocebus aethiops, Disease Models, Animal, Electron Transport Complex IV metabolism, Fibroblasts cytology, Fibroblasts enzymology, Heme Oxygenase (Decyclizing) genetics, Heme Oxygenase-1 genetics, Kidney cytology, Macrophages cytology, Membrane Proteins genetics, Mice, Oxidative Stress, Rats, Rats, Sprague-Dawley, Heme Oxygenase (Decyclizing) metabolism, Heme Oxygenase-1 metabolism, Kidney enzymology, Liver Diseases, Alcoholic enzymology, Macrophages enzymology, Membrane Proteins metabolism, Mitochondria enzymology
- Abstract
The inducible form of Heme Oxygenase-1 (HO-1), a major endoplasmic reticulum (ER) associated heme protein, is known to play important roles in protection against oxidative and chemical stress by degrading free heme released from degradation of heme proteins. In this study we show that induced expression of HO-1 by subjecting macrophage RAW-264.7 cells to chemical or physiological hypoxia resulted in significant translocation of HO-1 protein to mitochondria. Transient transfection of COS-7 cells with cloned cDNA also resulted in mitochondrial translocation of HO-1. Deletion of N-terminal ER targeting domain increased mitochondrial translocation under the transient transfection conditions. Mitochondrial localization of both intact HO-1 and N-terminal truncated HO-1 caused loss of heme aa-3 and cytochrome c oxidase (CcO) activity in COS-7 cells. The truncated protein, which localizes to mitochondria at higher levels, induced substantially steeper loss of CcO activity and reduced heme aa3 content. Furthermore, cells expressing mitochondria targeted HO-1 also induced higher ROS production. Consistent with dysfunctional state of mitochondria induced by HO-1, the mitochondrial recruitment of autophagy markers LC-3 and Drp-1 was also increased in these cells. Chronic ethanol feeding in rats also caused an increase in mitochondrial HO-1 and decrease in CcO activity. These results show that as opposed to the protective effect of the ER associated HO-1, mitochondria targeted HO-1 under normoxic conditions induces mitochondrial dysfunction.
- Published
- 2013
- Full Text
- View/download PDF
37. Smoke carcinogens cause bone loss through the aryl hydrocarbon receptor and induction of Cyp1 enzymes.
- Author
-
Iqbal J, Sun L, Cao J, Yuen T, Lu P, Bab I, Leu NA, Srinivasan S, Wagage S, Hunter CA, Nebert DW, Zaidi M, and Avadhani NG
- Subjects
- Animals, Aryl Hydrocarbon Hydroxylases deficiency, Aryl Hydrocarbon Hydroxylases genetics, Benzo(a)pyrene toxicity, Bone Resorption pathology, Cytochrome P-450 CYP1A1 biosynthesis, Cytochrome P-450 CYP1A1 deficiency, Cytochrome P-450 CYP1A1 genetics, Cytochrome P-450 CYP1A2 biosynthesis, Cytochrome P-450 CYP1A2 deficiency, Cytochrome P-450 CYP1A2 genetics, Cytochrome P-450 CYP1B1, Disease Models, Animal, Enzyme Induction genetics, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Polychlorinated Dibenzodioxins toxicity, Receptors, Aryl Hydrocarbon deficiency, Receptors, Aryl Hydrocarbon genetics, Smoking adverse effects, Smoking genetics, Nicotiana toxicity, Aryl Hydrocarbon Hydroxylases biosynthesis, Bone Resorption etiology, Bone Resorption metabolism, Carcinogens toxicity, Receptors, Aryl Hydrocarbon physiology, Smoke adverse effects
- Abstract
Smoking is a major risk factor for osteoporosis and fracture, but the mechanism through which smoke causes bone loss remains unclear. Here, we show that the smoke toxins benzo(a)pyrene (BaP) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) interact with the aryl hydrocarbon receptor (Ahr) to induce osteoclastic bone resorption through the activation of cytochrome P450 1a/1b (Cyp1) enzymes. BaP and TCDD enhanced osteoclast formation in bone marrow cell cultures and gavage with BaP stimulated bone resorption and osteoclastogenesis in vivo. The osteoclastogenesis triggered by BaP or RANK-L was reduced in Ahr(-/-) cells, consistent with the high bone mass noted in Ahr(-/-) male mice. The receptor activator of NF-κB ligand (RANK-L) also failed to induce the expression of Cyp1 enzymes in Ahr(-/-) cells. Furthermore, the osteoclastogenesis induced by TCDD was lower in Cyp1a1/1a2(-/-) and Cyp1a1/1a2/1b1(-/-) cultures, indicating that Ahr was upstream of the Cyp enzymes. Likewise, the pharmacological inhibition of the Cyp1 enzymes with tetramethylsilane or proadifen reduced osteoclastogenesis. Finally, deletion of the Cyp1a1, Cyp1a2, and Cyp1b1 in triple knockout mice resulted in reduced bone resorption and recapitulated the high bone mass phenotype of Ahr(-/-) mice. Overall, the data identify the Ahr and Cyp1 enzymes not only in the pathophysiology of smoke-induced osteoporosis, but also as potential targets for selective modulation by new therapeutics.
- Published
- 2013
- Full Text
- View/download PDF
38. Human cytochrome P450 2E1 mutations that alter mitochondrial targeting efficiency and susceptibility to ethanol-induced toxicity in cellular models.
- Author
-
Bansal S, Anandatheerthavarada HK, Prabu GK, Milne GL, Martin MV, Guengerich FP, and Avadhani NG
- Subjects
- Amino Acid Substitution, Animals, COS Cells, Central Nervous System Depressants pharmacology, Chlorocebus aethiops, Cytochrome P-450 CYP2E1 genetics, Ethanol pharmacology, Hep G2 Cells, Humans, Liver pathology, Mitochondria genetics, Mitochondria pathology, Mitochondrial Proteins genetics, Oxygen Consumption drug effects, Oxygen Consumption genetics, Protein Transport drug effects, Protein Transport genetics, Central Nervous System Depressants adverse effects, Cytochrome P-450 CYP2E1 metabolism, Ethanol adverse effects, Liver enzymology, Mitochondria enzymology, Mitochondrial Proteins metabolism, Models, Biological, Mutation, Missense
- Abstract
Human polymorphisms in the 5'-upstream regulatory regions and also protein coding regions of cytochrome P450 2E1 (CYP2E1) are known to be associated with several diseases, including cancer and alcohol liver toxicity. In this study, we report novel mutations in the N-terminal protein targeting regions of CYP2E1 that markedly affect subcellular localization of the protein. Variant W23R/W30R protein (termed W23/30R) is preferentially targeted to mitochondria but very poorly to the endoplasmic reticulum, whereas the L32N protein is preferentially targeted to the endoplasmic reticulum and poorly to mitochondria. These results explain the physiological significance of bimodal CYP targeting to the endoplasmic reticulum and mitochondria previously described. COS-7 cells and HepG2 cells stably expressing W23/30R mutations showed markedly increased alcohol toxicity in terms of increased production of reactive oxygen species, respiratory dysfunction, and loss of cytochrome c oxidase subunits and activity. Stable cells expressing the L32N variant, on the other hand, were relatively less responsive to alcohol-induced toxicity and mitochondrial dysfunction. These results further support our previous data, based on mutational studies involving altered targeting, indicating that mitochondria-targeted CYP2E1 plays an important role in alcohol liver toxicity. The results also provide an interesting new link to genetic variations affecting subcellular distribution of CYP2E1 with alcohol-induced toxicity.
- Published
- 2013
- Full Text
- View/download PDF
39. Metabolism of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine by mitochondrion-targeted cytochrome P450 2D6: implications in Parkinson disease.
- Author
-
Bajpai P, Sangar MC, Singh S, Tang W, Bansal S, Chowdhury G, Cheng Q, Fang JK, Martin MV, Guengerich FP, and Avadhani NG
- Subjects
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine adverse effects, 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine pharmacology, Adrenergic alpha-Antagonists pharmacology, Animals, Cell Line, Cytochrome P-450 CYP2D6 genetics, Dopamine Agents adverse effects, Dopamine Agents pharmacology, Dopaminergic Neurons pathology, Dynamins genetics, Dynamins metabolism, Humans, Mice, Mitochondria genetics, Mitochondrial Proteins genetics, Parkinsonian Disorders drug therapy, Parkinsonian Disorders genetics, Parkinsonian Disorders pathology, Quinidine pharmacology, Reactive Oxygen Species metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine pharmacokinetics, Cytochrome P-450 CYP2D6 metabolism, Dopamine Agents pharmacokinetics, Dopaminergic Neurons enzymology, Mitochondria metabolism, Mitochondrial Proteins metabolism, Parkinsonian Disorders enzymology
- Abstract
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a neurotoxic side product formed in the chemical synthesis of desmethylprodine opioid analgesic, which induces Parkinson disease. Monoamine oxidase B, present in the mitochondrial outer membrane of glial cells, catalyzes the oxidation of MPTP to the toxic 1-methyl-4-phenylpyridinium ion (MPP(+)), which then targets the dopaminergic neurons causing neuronal death. Here, we demonstrate that mitochondrion-targeted human cytochrome P450 2D6 (CYP2D6), supported by mitochondrial adrenodoxin and adrenodoxin reductase, can efficiently catalyze the metabolism of MPTP to MPP(+), as shown with purified enzymes and also in cells expressing mitochondrial CYP2D6. Neuro-2A cells stably expressing predominantly mitochondrion-targeted CYP2D6 were more sensitive to MPTP-mediated mitochondrial respiratory dysfunction and complex I inhibition than cells expressing predominantly endoplasmic reticulum-targeted CYP2D6. Mitochondrial CYP2D6 expressing Neuro-2A cells produced higher levels of reactive oxygen species and showed abnormal mitochondrial structures. MPTP treatment also induced mitochondrial translocation of an autophagic marker, Parkin, and a mitochondrial fission marker, Drp1, in differentiated neurons expressing mitochondrial CYP2D6. MPTP-mediated toxicity in primary dopaminergic neurons was attenuated by CYP2D6 inhibitor, quinidine, and also partly by monoamine oxidase B inhibitors deprenyl and pargyline. These studies show for the first time that dopaminergic neurons expressing mitochondrial CYP2D6 are fully capable of activating the pro-neurotoxin MPTP and inducing neuronal damage, which is effectively prevented by the CYP2D6 inhibitor quinidine.
- Published
- 2013
- Full Text
- View/download PDF
40. Mitochondrial dysfunction in myocardium obtained from clinically normal dogs, clinically normal anesthetized dogs, and dogs with dilated cardiomyopathy.
- Author
-
Sleeper MM, Rosato BP, Bansal S, and Avadhani NG
- Subjects
- Anesthesia, Inhalation, Anesthetics, Inhalation, Animals, Cardiomyopathy, Dilated pathology, Dogs, Mitochondria, Heart metabolism, Myocardium metabolism, Cardiomyopathy, Dilated veterinary, Dog Diseases pathology, Mitochondria, Heart pathology, Myocardium pathology
- Abstract
Objective: To compare mitochondrial complex I and complex IV activity in myocardial mitochondria of clinically normal dogs, clinically normal dogs exposed to inhalation anesthesia, and dogs affected with dilated cardiomyopathy., Sample: Myocardial samples obtained from 21 euthanized dogs (6 clinically normal [control] dogs, 5 clinically normal dogs subjected to inhalation anesthesia with isoflurane prior to euthanasia, 5 dogs with juvenile-onset dilated cardiomyopathy, and 5 dogs with adult-onset dilated cardiomyopathy)., Procedures: Activity of mitochondrial complex I and complex IV was assayed spectrophotometrically in isolated mitochondria from left ventricular tissue obtained from the 4 groups of dogs., Results: Activity of complex I and complex IV was significantly decreased in anesthetized dogs, compared with activities in the control dogs and dogs with juvenile-onset or adult-onset dilated cardiomyopathy., Conclusions and Clinical Relevance: Inhalation anesthesia disrupted the electron transport chain in the dogs, which potentially led to an outburst of reactive oxygen species that caused mitochondrial dysfunction. Inhalation anesthesia depressed mitochondrial function in dogs, similar to results reported in other species. This effect is important to consider when anesthetizing animals with myocardial disease and suggested that antioxidant treatments may be beneficial in some animals. Additionally, this effect should be considered when designing studies in which mitochondrial enzyme activity will be measured. Additional studies that include a larger number of animals are warranted.
- Published
- 2012
- Full Text
- View/download PDF
41. Cytochrome c oxidase dysfunction in oxidative stress.
- Author
-
Srinivasan S and Avadhani NG
- Subjects
- Animals, Apoptosis, Electron Transport Complex IV genetics, Electron Transport Complex IV metabolism, Enzyme Stability, Humans, Membrane Potential, Mitochondrial, Mitochondria enzymology, Mitochondria metabolism, Mitochondria physiology, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Subunits genetics, Protein Subunits metabolism, Reactive Oxygen Species metabolism, Electron Transport Complex IV physiology, Oxidative Stress
- Abstract
Cytochrome c oxidase (CcO) is the terminal oxidase of the mitochondrial electron transport chain. This bigenomic enzyme in mammals contains 13 subunits of which the 3 catalytic subunits are encoded by the mitochondrial genes. The remaining 10 subunits with suspected roles in the regulation, and/or assembly, are coded by the nuclear genome. The enzyme contains two heme groups (heme a and a3) and two Cu(2+) centers (Cu(2+) A and Cu(2+) B) as catalytic centers and handles more than 90% of molecular O(2) respired by the mammalian cells and tissues. CcO is a highly regulated enzyme which is believed to be the pacesetter for mitochondrial oxidative metabolism and ATP synthesis. The structure and function of the enzyme are affected in a wide variety of diseases including cancer, neurodegenerative diseases, myocardial ischemia/reperfusion, bone and skeletal diseases, and diabetes. Despite handling a high O(2) load the role of CcO in the production of reactive oxygen species still remains a subject of debate. However, a volume of evidence suggests that CcO dysfunction is invariably associated with increased mitochondrial reactive oxygen species production and cellular toxicity. In this paper we review the literature on mechanisms of multimodal regulation of CcO activity by a wide spectrum of physiological and pathological factors. We also review an array of literature on the direct or indirect roles of CcO in reactive oxygen species production., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
42. Silencing of IkBβ mRNA causes disruption of mitochondrial retrograde signaling and suppression of tumor growth in vivo.
- Author
-
Tang W, Chowdhury AR, Guha M, Huang L, Van Winkle T, Rustgi AK, and Avadhani NG
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, DNA, Mitochondrial genetics, Energy Metabolism, Gene Silencing, Humans, I-kappa B Proteins antagonists & inhibitors, I-kappa B Proteins genetics, Ki-67 Antigen analysis, Mice, Mitochondria physiology, NF-kappa B physiology, Neoplasms pathology, Neoplasms prevention & control, RNA, Small Interfering genetics, DNA, Mitochondrial physiology, I-kappa B Proteins physiology, Neoplasms etiology, Signal Transduction physiology
- Abstract
A number of studies show that mitochondrial DNA (mtDNA) depletion and attendant activation of retrograde signaling induces tumor progression. We have reported previously that activation of a novel nuclear factor-Kappa B pathway is critical for the propagation of mitochondrial retrograde signaling, which induces both phenotypic and morphological changes in C2C12 myoblasts and A549 lung carcinoma cells. In this study, we investigated the role of stress-induced nuclear factor-Kappa B in tumor progression in xenotransplanted mice. We used a retroviral system for the inducible expression of small interfering RNA against IkBα and IkBβ mRNAs. Expression of small interfering RNA against IkBβ markedly impaired tumor growth and invasive ability of mtDNA-depleted C2C12 myoblasts and also thwarted anchorage-independent growth of the cells. Knockdown of IkBα mRNA, however, did not have any modulatory effect in this cell system. Moreover, expression of small interfering RNA against IkBβ reduced the expression of marker genes for retrograde signaling and tumor growth in xenografts of mtDNA-depleted cells. Our findings demonstrate that IkBβ is a master regulator of mitochondrial retrograde signaling pathway and that the retrograde signaling plays a role in tumor growth in vivo. In this regard, IkBβ supports the tumorigenic potential of mtDNA-depleted C2C12 cells.
- Published
- 2012
- Full Text
- View/download PDF
43. Additive effects of mitochondrion-targeted cytochrome CYP2E1 and alcohol toxicity on cytochrome c oxidase function and stability of respirosome complexes.
- Author
-
Bansal S, Srinivasan S, Anandasadagopan S, Chowdhury AR, Selvaraj V, Kalyanaraman B, Joseph J, and Avadhani NG
- Subjects
- Animals, Antioxidants pharmacology, COS Cells, Central Nervous System Depressants toxicity, Chlorocebus aethiops, DNA, Mitochondrial genetics, DNA, Mitochondrial metabolism, Electron Transport Complex IV genetics, Hep G2 Cells, Humans, Immunoblotting, Liver drug effects, Liver metabolism, Liver pathology, Microsomes drug effects, Microsomes metabolism, Mitochondria genetics, Mitochondria metabolism, Mitochondria, Liver drug effects, Mitochondria, Liver genetics, Mitochondria, Liver metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Oxygen Consumption drug effects, Protein Carbonylation drug effects, Rats, Rats, Sprague-Dawley, Reactive Oxygen Species metabolism, Reverse Transcriptase Polymerase Chain Reaction, Transcription, Genetic drug effects, Cytochrome P-450 CYP2E1 metabolism, Electron Transport drug effects, Electron Transport Complex IV metabolism, Ethanol toxicity, Mitochondria drug effects
- Abstract
Alcohol treatment induces oxidative stress by a combination of increased production of partially reduced oxygen species and decreased cellular antioxidant pool, including GSH. Recently, we showed that mitochondrion-targeted CYP2E1 augments alcohol-mediated toxicity, causing an increase in reactive oxygen species production and oxidative stress. Here, we show that cytochrome c oxidase (CcO), the terminal oxidase of the mitochondrial respiratory chain, is a critical target of CYP2E1-mediated alcohol toxicity. COS-7 and Hep G2 cell lines expressing predominantly mitochondrion-targeted (Mt(++)) CYP2E1 and livers from alcohol-treated rats showed loss of CcO activity and increased protein carbonylation, which was accompanied by a decline in the steady state levels of subunits I, IVI1, and Vb of the CcO complex. This was also accompanied by reduced mitochondrial DNA content and reduced mitochondrial mRNA. These changes were more prominent in Mt(++) cells in comparison with wild type (WT) CYP2E1-expressing or ER(+) (mostly microsome-targeted) cells. In addition, mitochondrion-specific antioxidants, ubiquinol conjugated to triphenyl phosphonium, triphenylphosphonium conjugated carboxyl proxyl, and the CYP2E1 inhibitor diallyl sulfide prevented the loss of CcO activity and the CcO subunits, most likely through reduced oxidative damage to the enzyme complex. Our results suggest that damage to CcO and dissociation of respirosome complexes are critical factors in alcohol-induced toxicity, which is augmented by mitochondrion-targeted CYP2E1. We propose that CcO is one of the direct and immediate targets of alcohol-induced toxicity causing respiratory dysfunction.
- Published
- 2012
- Full Text
- View/download PDF
44. The effects of smoke carcinogens on bone.
- Author
-
Yan C, Avadhani NG, and Iqbal J
- Subjects
- Bone Density drug effects, Bone Density physiology, Bone Remodeling drug effects, Bone Remodeling physiology, Bone and Bones physiopathology, Humans, Osteoporosis epidemiology, Osteoporosis physiopathology, Risk Factors, Bone and Bones drug effects, Carcinogens pharmacology, Smoking adverse effects
- Abstract
The greatest cause of preventable morbidity and mortality is smoking, and one of the often-underappreciated effects of smoking is profound bone loss. The existing clinical paradigm for smoking is that there is a low turnover osteoporosis. This review highlights findings from recent clinical trials and animal research demonstrating either support or conflict with the existing paradigm. Clinically, it is noted that markers of bone formation are often normal in smokers; these clinical findings conflict with well-conducted animal research demonstrating that carcinogens acting on the aryl hydrogen receptor can significantly reduce osteoblast formation and function. Regarding bone resorption, highlights from recent clinical studies suggest that bone remodeling is increased in smokers. Directly contradicting this enhanced osteoclastogenesis are several animal studies all demonstrating significant inhibition of osteoclast formation and function upon exposure to smoke carcinogens. Future research is needed to clarify whether smoking is truly a low bone remodeling osteoporosis, or an osteoclast-driven bone destruction, with inappropriately normal bone formation.
- Published
- 2011
- Full Text
- View/download PDF
45. Targeting of the same proteins to multiple subcellular destinations: mechanisms and physiological implications.
- Author
-
Avadhani NG
- Subjects
- Animals, Humans, Protein Transport, Subcellular Fractions, Cell Physiological Phenomena, Proteins metabolism
- Published
- 2011
- Full Text
- View/download PDF
46. Bimodal targeting of cytochrome P450s to endoplasmic reticulum and mitochondria: the concept of chimeric signals.
- Author
-
Avadhani NG, Sangar MC, Bansal S, and Bajpai P
- Subjects
- Amino Acid Sequence, Animals, Humans, Molecular Sequence Data, Protein Transport, Sequence Homology, Amino Acid, Cytochrome P-450 Enzyme System metabolism, Endoplasmic Reticulum metabolism, Mitochondria metabolism, Signal Transduction
- Abstract
Targeting signals are critical for proteins to find their specific cellular destination. Signals for protein targeting to the endoplasmic reticulum (ER), mitochondria, peroxisome and nucleus are distinct and the mechanisms of protein translocation across these membrane compartments also vary markedly. Recently, however, a number of proteins have been shown to be present in multiple cellular sites such as mitochondria and ER, cytosol and mitochondria, plasma membrane and mitochondria, and peroxisome and mitochondria suggesting the occurrence of multimodal targeting signals in some cases. Cytochrome P450 monooxygenases (CYPs), which play crucial roles in pharmacokinetics and pharmacodynamics of drugs and toxins, are the prototype of bimodally targeted proteins. Several members of family 1, 2 and 3 CYPs have now been reported to be associated with mitochondria and plasma membrane in addition to the ER. This review highlights the mechanisms of bimodal targeting of CYP1A1, 2B1, 2E1 and 2D6 to mitochondria and ER. The bimodal targeting of these proteins is driven by their N-terminal signals which carry essential elements of both ER targeting and mitochondria targeting signals. These multimodal signals have been termed chimeric signals appropriately to describe their dual targeting property. The cryptic mitochondrial targeting signals of CYP2B1, 2D6, 2E1 require activation by protein kinase A or protein kinase C mediated phosphorylation at sites immediately flanking the targeting signal and/or membrane anchoring regions. The cryptic mitochondria targeting signal of CYP1A1 requires activation by endoproteolytic cleavage by a cytosolic endoprotease, which exposes the mitochondrial signal. This review discusses both mechanisms of bimodal targeting and toxicological consequences of mitochondria targeted CYP proteins., (© 2011 The Authors Journal compilation © 2011 FEBS.)
- Published
- 2011
- Full Text
- View/download PDF
47. Impaired mitochondrial respiratory functions and oxidative stress in streptozotocin-induced diabetic rats.
- Author
-
Raza H, Prabu SK, John A, and Avadhani NG
- Subjects
- Aconitate Hydratase metabolism, Animals, Diabetes Mellitus, Experimental metabolism, Male, Mitochondria metabolism, Rats, Sprague-Dawley, Reactive Oxygen Species metabolism, Streptozocin, Diabetes Mellitus, Experimental physiopathology, Mitochondria physiology, Oxidative Stress
- Abstract
We have previously shown a tissue-specific increase in oxidative stress in the early stages of streptozotocin (STZ)-induced diabetic rats. In this study, we investigated oxidative stress-related long-term complications and mitochondrial dysfunctions in the different tissues of STZ-induced diabetic rats (>15 mM blood glucose for 8 weeks). These animals showed a persistent increase in reactive oxygen and nitrogen species (ROS and RNS, respectively) production. Oxidative protein carbonylation was also increased with the maximum effect observed in the pancreas of diabetic rats. The activities of mitochondrial respiratory enzymes ubiquinol: cytochrome c oxidoreductase (Complex III) and cytochrome c oxidase (Complex IV) were significantly decreased while that of NADH:ubiquinone oxidoreductase (Complex I) and succinate:ubiquinone oxidoreductase (Complex II) were moderately increased in diabetic rats, which was confirmed by the increased expression of the 70 kDa Complex II sub-unit. Mitochondrial matrix aconitase, a ROS sensitive enzyme, was markedly inhibited in the diabetic rat tissues. Increased expression of oxidative stress marker proteins Hsp-70 and HO-1 was also observed along with increased expression of nitric oxide synthase. These results suggest that mitochondrial respiratory complexes may play a critical role in ROS/RNS homeostasis and oxidative stress related changes in type 1 diabetes and may have implications in the etiology of diabetes and its complications.
- Published
- 2011
- Full Text
- View/download PDF
48. Cell signaling.
- Author
-
Iqbal J, Zaidi M, and Avadhani NG
- Subjects
- Animals, Extracellular Fluid physiology, Gene Expression Regulation physiology, Humans, Intracellular Fluid physiology, RNA, Messenger physiology, Cell Physiological Phenomena physiology, Signal Transduction physiology
- Abstract
This review explores advances in our understanding of dynamicism in cellular signaling. Areas highlighted include the role of stochasticity in producing diversity in analogous signaling circumstances; population desynchronization's effect in masking newly appreciated repetitive bursts in protein phosphorylation and messenger RNA production; double-positive feedback interactions and their ability to synchronize multiple signal transduction pathways; scaffolding proteins control over signaling feedback; and frequency-responsive transcriptional regulation as an example of dynamicism in signaling., (© 2010 New York Academy of Sciences.)
- Published
- 2010
- Full Text
- View/download PDF
49. Activation of Akt is essential for the propagation of mitochondrial respiratory stress signaling and activation of the transcriptional coactivator heterogeneous ribonucleoprotein A2.
- Author
-
Guha M, Fang JK, Monks R, Birnbaum MJ, and Avadhani NG
- Subjects
- Animals, Apoptosis, Cathepsin L metabolism, Cell Line, Cell Nucleus enzymology, Cell Respiration, DNA, Mitochondrial metabolism, Enzyme Activation, Gene Silencing, Humans, Mice, Mitochondria genetics, Models, Biological, Phenotype, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Promoter Regions, Genetic genetics, Protein Binding, Protein Transport, Receptor, IGF Type 1 metabolism, Transcriptional Activation genetics, Heterogeneous-Nuclear Ribonucleoprotein Group A-B metabolism, Mitochondria enzymology, Mitochondria pathology, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Stress, Physiological, Trans-Activators metabolism
- Abstract
Mitochondrial respiratory stress (also called mitochondrial retrograde signaling) activates a Ca(2+)/calcineurin-mediated signal that culminates in transcription activation/repression of a large number of nuclear genes. This signal is propagated through activation of the regulatory proteins NFκB c-Rel/p50, C/EBPδ, CREB, and NFAT. Additionally, the heterogeneous ribonucleoprotein A2 (hnRNPA2) functions as a coactivator in up-regulating the transcription of Cathepsin L, RyR1, and Glut-4, the target genes of stress signaling. Activation of IGF1R, which causes a metabolic switch to glycolysis, cell invasiveness, and resistance to apoptosis, is a phenotypic hallmark of C2C12 myoblasts subjected to mitochondrial stress. In this study, we report that mitochondrial stress leads to increased expression, activation, and nuclear localization of Akt1. Mitochondrial respiratory stress also activates Akt1-gene expression, which involves hnRNPA2 as a coactivator, indicating a complex interdependency of these two factors. Using Akt1(-/-) mouse embryonic fibroblasts and Akt1 mRNA-silenced C2C12 cells, we show that Akt1-mediated phosphorylation is crucial for the activation and recruitment of hnRNPA2 to the enhanceosome complex. Akt1 mRNA silencing in mtDNA-depleted cells resulted in reversal of the invasive phenotype, accompanied by sensitivity to apoptotic stimuli. These results show that Akt1 is an important regulator of the nuclear transcriptional response to mitochondrial stress.
- Published
- 2010
- Full Text
- View/download PDF
50. Bimodal targeting of microsomal cytochrome P450s to mitochondria: implications in drug metabolism and toxicity.
- Author
-
Sangar MC, Bansal S, and Avadhani NG
- Subjects
- Animals, Drug-Related Side Effects and Adverse Reactions, Endoplasmic Reticulum enzymology, Humans, Microsomes metabolism, Oxidative Stress, Rodentia, Cytochrome P-450 Enzyme System metabolism, Mitochondria enzymology, Pharmaceutical Preparations metabolism
- Abstract
Importance of the Field: Microsomal CYPs are critical for drug metabolism and toxicity. Recent studies show that these CYPs are also present in the mitochondrial compartment of human and rodent tissues. Mitochondrial CYP1A1 and 2E1 show both overlapping and distinct metabolic activities compared to microsomal forms. Mitochondrial CYP2E1 also induces oxidative stress. The mechanisms of mitochondria targeting of CYPs and their role in drug metabolism and toxicity are important factors to consider while determining the drug dose and in drug development., Areas Covered in This Review: This review highlights the mechanisms of bimodal targeting of CYP1A1, 2B1, 2E1 and 2D6 to mitochondria and microsomes. The review also discusses differences in structure and function of mitochondrial CYPs., What the Readers Will Gain: A comprehensive review of the literature on drug metabolism in the mitochondrial compartment and their potential for inducing mitochondrial dysfunction., Take Home Message: Studies on the biochemistry, pharmacology and pharmacogenetic analysis of CYPs are mostly focused on the molecular forms associated with the microsomal membrane. However, the mitochondrial CYPs in some individuals can represent a substantial part of the tissue pool and contribute in a significant way to drug metabolism, clearance and toxicity.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.