Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
PLoS One ; 15(9): e0239625, 2020.
Article in English | MEDLINE | ID: mdl-32966340

ABSTRACT

During alcohol consumption, the esophageal mucosa is directly exposed to high concentrations of ethanol (EtOH). We therefore investigated the response of normal human esophageal epithelial cell lines EPC1, EPC2 and EPC3 to acute EtOH exposure. While these cells were able to tolerate 2% EtOH for 8 h in both three-dimensional organoids and monolayer culture conditions, RNA sequencing suggested that EtOH induced mitochondrial dysfunction. With EtOH treatment, EPC1 and EPC2 cells also demonstrated decreased mitochondrial ATPB protein expression by immunofluorescence and swollen mitochondria lacking intact cristae by transmission electron microscopy. Mitochondrial membrane potential (ΔΨm) was decreased in a subset of EPC1 and EPC2 cells stained with ΔΨm-sensitive dye MitoTracker Deep Red. In EPC2, EtOH decreased ATP level while impairing mitochondrial respiration and electron transportation chain functions, as determined by ATP fluorometric assay, respirometry, and liquid chromatography-mass spectrometry. Additionally, EPC2 cells demonstrated enhanced oxidative stress by flow cytometry for mitochondrial superoxide (MitoSOX), which was antagonized by the mitochondria-specific antioxidant MitoCP. Concurrently, EPC1 and EPC2 cells underwent autophagy following EtOH exposure, as evidenced by flow cytometry for Cyto-ID, which detects autophagic vesicles, and immunoblots demonstrating induction of the lipidated and cleaved form of LC3B and downregulation of SQSTM1/p62. In EPC1 and EPC2, pharmacological inhibition of autophagy flux by chloroquine increased mitochondrial oxidative stress while decreasing cell viability. In EPC2, autophagy induction was coupled with phosphorylation of AMP activated protein kinase (AMPK), a cellular energy sensor responding to low ATP levels, and dephosphorylation of downstream substrates of mechanistic Target of Rapamycin Complex (mTORC)-1 signaling. Pharmacological AMPK activation by AICAR decreased EtOH-induced reduction of ΔΨm and ATP in EPC2. Taken together, acute EtOH exposure leads to mitochondrial dysfunction and oxidative stress in esophageal keratinocytes, where the AMPK-mTORC1 axis may serve as a regulatory mechanism to activate autophagy to provide cytoprotection against EtOH-induced cell injury.


Subject(s)
Autophagy , Esophagus/cytology , Keratinocytes/metabolism , Mitochondria/metabolism , Oxidative Stress , AMP-Activated Protein Kinase Kinases , Animals , Cell Line , Cells, Cultured , Ethanol/pharmacology , Female , Keratinocytes/drug effects , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
3.
Oncotarget ; 10(58): 6245-6259, 2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31692873

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.

4.
FASEB J ; 33(8): 9167-9181, 2019 08.
Article in English | MEDLINE | ID: mdl-31063702

ABSTRACT

The mitochondria-to-nucleus retrograde signaling (MtRS) pathway aids in cellular adaptation to stress. We earlier reported that the Ca2+- 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.


Subject(s)
Electron Transport Complex IV/metabolism , Macrophages/cytology , Macrophages/physiology , Osteoclasts/cytology , Osteoclasts/physiology , Phagocytosis/physiology , 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
6.
Oncogene ; 38(12): 2092-2107, 2019 03.
Article in English | MEDLINE | ID: mdl-30442981

ABSTRACT

Aberrant Notch signaling is implicated in several cancers, including breast cancer. However, the mechanistic details of the specific receptors and function of ligand-mediated Notch signaling that promote breast cancer remains elusive. In our studies we show that DLL1, a Notch signaling ligand, is significantly overexpressed in ERα+ luminal breast cancer. Intriguingly, DLL1 overexpression correlates with poor prognosis in ERα+ luminal breast cancer, but not in other subtypes of breast cancer. In addition, this effect is specific to DLL1, as other Notch ligands (DLL3, JAGGED1, and JAGGED2) do not influence the clinical outcome of ERα+ patients. Genetic studies show that DLL1-mediated Notch signaling in breast cancer is important for tumor cell proliferation, angiogenesis, and cancer stem cell function. Consistent with prognostic clinical data, we found the tumor-promoting function of DLL1 is exclusive to ERα+ luminal breast cancer, as loss of DLL1 inhibits both tumor growth and lung metastasis of luminal breast cancer. Importantly, we find that estrogen signaling stabilizes DLL1 protein by preventing its proteasomal and lysososmal degradations. Moreover, estrogen inhibits ubiquitination of DLL1. Together, our results highlight an unexpected and novel subtype-specific function of DLL1 in promoting luminal breast cancer that is regulated by estrogen signaling. Our studies also emphasize the critical role of assessing subtype-specific mechanisms driving tumor growth and metastasis to generate effective subtype-specific therapeutics.


Subject(s)
Breast Neoplasms/pathology , Estrogens/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Calcium-Binding Proteins , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Disease Progression , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lysosomes/metabolism , Mice , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic , Prognosis , Ubiquitination
7.
PLoS One ; 13(12): e0209489, 2018.
Article in English | MEDLINE | ID: mdl-30576337

ABSTRACT

Osteosarcoma (OSA) is an aggressive mesenchymal tumor of the bone that affects children and occurs spontaneously in dogs. Human and canine OSA share similar clinical, biological and genetic features, which make dogs an excellent comparative model to investigate the etiology and pathogenesis of OSA. Mitochondrial (mt) defects have been reported in many different cancers including OSA, although it is not known whether these defects contribute to OSA progression and metastasis. Taking a comparative approach using canine OSA cell lines and tumor tissues we investigated the effects of mtDNA content and dysfunction on OSA biology. OSA tumor tissues had low mtDNA contents compared to the matched non-tumor tissues. We observed mitochondrial heterogeneity among the OSA cell lines and the most invasive cells expressing increased levels of OSA metastasis genes contained the highest amount of mitochondrial defects (reduced mtDNA copies, mt respiration, and expression of electron transport chain proteins). While mitochondria maintain a filamentous network in healthy cells, the mitochondrial morphology in OSA cells were mostly "donut shaped", typical of "stressed" mitochondria. Moreover the expression levels of mitochondrial retrograde signaling proteins Akt1, IGF1R, hnRNPA2 and NFkB correlated with the invasiveness of the OSA cells. Furthermore, we demonstrate the causal role of mitochondrial defects in inducing the invasive phenotype by Ethidium Bromide induced-mtDNA depletion in OSA cells. Our data suggest that defects in mitochondrial genome and function are prevalent in OSA and that lower mtDNA content is associated with higher tumor cell invasiveness. We propose that mt defects in OSA might serve as a prognostic biomarker and a target for therapeutic intervention in OSA patients.


Subject(s)
Bone Neoplasms/genetics , Dog Diseases/genetics , Genome, Mitochondrial , Osteosarcoma/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/isolation & purification , Biomarkers, Tumor/metabolism , Biopsy , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Bone Neoplasms/veterinary , Cancer Vaccines/therapeutic use , Carboplatin/therapeutic use , Cell Line, Tumor , DNA, Mitochondrial/genetics , DNA, Mitochondrial/isolation & purification , DNA, Mitochondrial/metabolism , Dog Diseases/pathology , Dog Diseases/therapy , Dogs , Ethidium/toxicity , Mitochondria/drug effects , Mitochondria/genetics , Neoplasm Invasiveness/genetics , Osteosarcoma/pathology , Osteosarcoma/therapy , Osteosarcoma/veterinary , Prognosis , Treatment Outcome
8.
PLoS One ; 13(11): e0206897, 2018.
Article in English | MEDLINE | ID: mdl-30427907

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.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Histones/metabolism , Mitochondria/metabolism , Telomere Shortening/genetics , Telomere/metabolism , 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
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1060-1071, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29309924

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.


Subject(s)
DNA, Mitochondrial , Mitochondria , Mitochondrial Proteins , Neoplasm Proteins , RNA-Binding Proteins , Triple Negative Breast Neoplasms , Cell Line, Tumor , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Humans , 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
10.
Biochim Biophys Acta Bioenerg ; 1858(7): 519-528, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28442264

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.


Subject(s)
ATP-Dependent Proteases/metabolism , Cell Hypoxia/physiology , Electron Transport Complex IV/metabolism , Mitochondria, Heart/enzymology , Mitochondrial Proteins/metabolism , Myocardial Ischemia/enzymology , 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
11.
Biochim Biophys Acta Bioenerg ; 1858(8): 602-614, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28104365

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.


Subject(s)
Cell Transformation, Neoplastic , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Neoplasms/metabolism , 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
12.
Cell Discov ; 2: 16045, 2016.
Article in English | MEDLINE | ID: mdl-27990297

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.

13.
Mol Cell Oncol ; 3(2): e1085120, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27308608

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.

14.
Am J Cancer Res ; 6(4): 781-96, 2016.
Article in English | MEDLINE | ID: mdl-27186430

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.

15.
J Clin Invest ; 126(5): 1834-56, 2016 05 02.
Article in English | MEDLINE | ID: mdl-27043285

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.


Subject(s)
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 , 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
16.
Ann N Y Acad Sci ; 1364: 52-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25800988

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.


Subject(s)
Cathepsin K/metabolism , Mitochondria/metabolism , Osteoclasts/metabolism , Osteogenesis , Promoter Regions, Genetic , Signal Transduction , 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
17.
Genom Data ; 6: 99-107, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26697345

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.

18.
Cell Rep ; 11(5): 785-797, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25921537

ABSTRACT

Expression of type I interferons (IFNs) can be induced by DNA-damaging agents, but the mechanisms and significance of this regulation are not completely understood. We found that the transcription factor IRF3, activated in an ATM-IKKα/ß-dependent manner, stimulates cell-autonomous IFN-ß expression in response to double-stranded DNA breaks. Cells and tissues with accumulating DNA damage produce endogenous IFN-ß and stimulate IFN signaling in vitro and in vivo. In turn, IFN acts to amplify DNA-damage responses, activate the p53 pathway, promote senescence, and inhibit stem cell function in response to telomere shortening. Inactivation of the IFN pathway abrogates the development of diverse progeric phenotypes and extends the lifespan of Terc knockout mice. These data identify DNA-damage-response-induced IFN signaling as a critical mechanism that links accumulating DNA damage with senescence and premature aging.


Subject(s)
Cellular Senescence , DNA Damage , Interferon-beta/metabolism , Animals , Antibodies, Neutralizing/immunology , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line , DNA Damage/drug effects , Humans , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/antagonists & inhibitors , Interferon-beta/genetics , Intestinal Mucosa/metabolism , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , RNA Interference , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Telomerase/deficiency , Telomerase/genetics , Telomerase/metabolism , Telomere/metabolism , Tumor Suppressor Protein p53/metabolism
19.
Mitochondrion ; 13(6): 577-91, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24004957

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.


Subject(s)
Energy Metabolism , Epigenesis, Genetic , Mitochondria/metabolism , Neoplasms/metabolism , Signal Transduction , Humans , Neoplasms/genetics
20.
Carcinogenesis ; 33(9): 1762-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22637744

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 IkBß mRNAs. Expression of small interfering RNA against IkBß 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 IkBß reduced the expression of marker genes for retrograde signaling and tumor growth in xenografts of mtDNA-depleted cells. Our findings demonstrate that IkBß 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, IkBß supports the tumorigenic potential of mtDNA-depleted C2C12 cells.


Subject(s)
DNA, Mitochondrial/physiology , I-kappa B Proteins/physiology , Neoplasms/etiology , Signal Transduction/physiology , 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
SELECTION OF CITATIONS
SEARCH DETAIL
...