Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Nat Cell Biol ; 14(12): 1336-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23178883

ABSTRACT

Ca(2+) flux across the mitochondrial inner membrane regulates bioenergetics, cytoplasmic Ca(2+) signals and activation of cell death pathways. Mitochondrial Ca(2+) uptake occurs at regions of close apposition with intracellular Ca(2+) release sites, driven by the inner membrane voltage generated by oxidative phosphorylation and mediated by a Ca(2+) selective ion channel (MiCa; ref. ) called the uniporter whose complete molecular identity remains unknown. Mitochondrial calcium uniporter (MCU) was recently identified as the likely ion-conducting pore. In addition, MICU1 was identified as a mitochondrial regulator of uniporter-mediated Ca(2+) uptake in HeLa cells. Here we identified CCDC90A, hereafter referred to as MCUR1 (mitochondrial calcium uniporter regulator 1), an integral membrane protein required for MCU-dependent mitochondrial Ca(2+) uptake. MCUR1 binds to MCU and regulates ruthenium-red-sensitive MCU-dependent Ca(2+) uptake. MCUR1 knockdown does not alter MCU localization, but abrogates Ca(2+) uptake by energized mitochondria in intact and permeabilized cells. Ablation of MCUR1 disrupts oxidative phosphorylation, lowers cellular ATP and activates AMP kinase-dependent pro-survival autophagy. Thus, MCUR1 is a critical component of a mitochondrial uniporter channel complex required for mitochondrial Ca(2+) uptake and maintenance of normal cellular bioenergetics.


Subject(s)
Calcium/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Animals , COS Cells , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , HeLa Cells , Humans , Male , Membrane Proteins/genetics , Mice , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Proteins/genetics
3.
Mol Cell Biol ; 31(18): 3745-58, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21746883

ABSTRACT

Necroptosis represents a form of alternative programmed cell death that is dependent on the kinase RIP1. RIP1-dependent necroptotic death manifests as increased reactive oxygen species (ROS) production in mitochondria and is accompanied by loss of ATP biogenesis and eventual dissipation of mitochondrial membrane potential. Here, we show that tumor necrosis factor alpha (TNF-α)-induced necroptosis requires the adaptor proteins FADD and NEMO. FADD was found to mediate formation of the TNF-α-induced pronecrotic RIP1-RIP3 kinase complex, whereas the IκB Kinase (IKK) subunit NEMO appears to function downstream of RIP1-RIP3. Interestingly, loss of RelA potentiated TNF-α-dependent necroptosis, indicating that NEMO regulates necroptosis independently of NF-κB. Using both pharmacologic and genetic approaches, we demonstrate that the overexpression of antioxidants alleviates ROS elevation and necroptosis. Finally, elimination of BAX and BAK or overexpression of Bcl-x(L) protects cells from necroptosis at a later step. These findings provide evidence that mitochondria play an amplifying role in inflammation-induced necroptosis.


Subject(s)
Apoptosis/physiology , Fas-Associated Death Domain Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Necrosis/metabolism , Tumor Necrosis Factor-alpha/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Blotting, Western , Flow Cytometry , GTPase-Activating Proteins/metabolism , Gene Knockout Techniques , Immunoprecipitation , Membrane Potential, Mitochondrial , Mice , Mitochondria/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2-Associated X Protein/deficiency
4.
Mol Cell Biol ; 31(14): 2934-46, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21576359

ABSTRACT

Interferons (IFNs) are cytokines with well-described immunomodulatory and antiviral properties, but less is known about the mechanisms by which they promote cell survival or cell death. Here, we show that IFN-γ induces RIP1 kinase-dependent necroptosis in mammalian cells deficient in NF-κB signaling. Induction of necroptosis by IFN-γ was found to depend on Jak1 and partially on STAT1. We also demonstrate that IFN-γ activates IκB kinase ß (IKKß)-dependent NF-κB to regulate a transcriptional program that protects cells from necroptosis. IFN-γ induced progressive accumulation of reactive oxygen species (ROS) and eventual loss of mitochondrial membrane potential in cells lacking the NF-κB subunit RelA. Whole-genome microarray analyses identified sod2, encoding the antioxidant enzyme manganese superoxide dismutase (MnSOD), as a RelA target and potential antinecroptotic gene. Overexpression of MnSOD inhibited IFN-γ-mediated ROS accumulation and partially rescued RelA-deficient cells from necroptosis, while RNA interference (RNAi)-mediated silencing of sod2 expression increased susceptibility to IFN-γ-induced cell death. Together, these studies demonstrate that NF-κB protects cells from IFN-γ-mediated necroptosis by transcriptionally activating a survival response that quenches ROS to preserve mitochondrial integrity.


Subject(s)
Cell Death/drug effects , Interferon-gamma/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism , Animals , Cell Death/physiology , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , HEK293 Cells , HeLa Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Janus Kinase 1/metabolism , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Knockout , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factor RelA/genetics , Transcription, Genetic , Tumor Necrosis Factor-alpha/pharmacology
5.
J Cell Biol ; 190(3): 391-405, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20679432

ABSTRACT

Oxidant stress influences many cellular processes, including cell growth, differentiation, and cell death. A well-recognized link between these processes and oxidant stress is via alterations in Ca(2+) signaling. However, precisely how oxidants influence Ca(2+) signaling remains unclear. Oxidant stress led to a phenotypic shift in Ca(2+) mobilization from an oscillatory to a sustained elevated pattern via calcium release-activated calcium (CRAC)-mediated capacitive Ca(2+) entry, and stromal interaction molecule 1 (STIM1)- and Orai1-deficient cells are resistant to oxidant stress. Functionally, oxidant-induced Ca(2+) entry alters mitochondrial Ca(2+) handling and bioenergetics and triggers cell death. STIM1 is S-glutathionylated at cysteine 56 in response to oxidant stress and evokes constitutive Ca(2+) entry independent of intracellular Ca(2+) stores. These experiments reveal that cysteine 56 is a sensor for oxidant-dependent activation of STIM1 and demonstrate a molecular link between oxidant stress and Ca(2+) signaling via the CRAC channel.


Subject(s)
Glutathione/metabolism , Homeostasis , Membrane Proteins/metabolism , Mitochondria/metabolism , Animals , COS Cells , Cells, Cultured , Chickens , Chlorocebus aethiops , Humans , Membrane Proteins/deficiency
6.
Free Radic Biol Med ; 48(2): 306-17, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19897030

ABSTRACT

Nitric oxide (NO) and other reactive nitrogen species target multiple sites in the mitochondria to influence cellular bioenergetics and survival. Kinetic imaging studies revealed that NO from either activated macrophages or donor compounds rapidly diffuses to the mitochondria, causing a dose-dependent progressive increase in NO-dependent DAF fluorescence, which corresponded to mitochondrial membrane potential loss and initiated alterations in cellular bioenergetics that ultimately led to necrotic cell death. Cellular dysfunction is mediated by an elevated 3-nitrotyrosine signature of the mitochondrial complex I subunit NDUFB8, which is vital for normal mitochondrial function as evidenced by selective knockdown via siRNA. Overexpression of mitochondrial superoxide dismutase substantially decreased NDUFB8 nitration and restored mitochondrial homeostasis. Further, treatment of cells with either necrostatin-1 or siRNA knockdown of RIP1 and RIP3 prevented NO-mediated necrosis. This work demonstrates that the interaction between NO and mitochondrially derived superoxide alters mitochondrial bioenergetics and cell function, thus providing a molecular mechanism for reactive oxygen and nitrogen species-mediated alterations in mitochondrial homeostasis.


Subject(s)
Electron Transport Complex I/metabolism , Endothelium, Vascular/metabolism , Fibroblasts/metabolism , Macrophages/metabolism , Mitochondria/metabolism , Animals , Cell Line , Electron Transport Complex I/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Macrophages/drug effects , Macrophages/pathology , Membrane Potential, Mitochondrial , Mice , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/genetics , Necrosis/genetics , Nitric Oxide/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Protein Subunits/metabolism , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
7.
Mol Cell Biol ; 29(11): 3099-112, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19332555

ABSTRACT

Ethanol intoxication stimulates the production of proinflammatory cytokines, increases the formation of reactive oxygen species, and induces mitochondrial impairment. However, information is limited as to the exact sequence and components involved in ethanol-induced hepatotoxicity. Acute ethanol exposure enhances mitochondrial superoxide (O(2)(*-)) production and impairs mitochondrial Ca(2+) handling. In turn, O(2)(*-) facilitates cytochrome c release and mitochondrial membrane potential loss that is not dependent upon H(2)O(2) and divalent cations and requires Bak in a Bax-independent fashion. Furthermore, triggering of Bak's proapoptotic activity requires the cytosolic presence of Bid, a BH3-only protein that is processed by the initiator caspase-2. Together, these studies identify an O(2)(*-)-driven, caspase-initiated apoptotic pathway that selectively involves the Bcl-2 family proteins Bid and Bak. This pathway manifests itself during chronic ethanol consumption in aged animals and identifies caspase-2, Bid, and Bak as essential mediators of O(2)(*-)-induced apoptosis that may prove effective targets for the development of therapeutics to treat alcoholic liver disease.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Superoxides/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Aging/drug effects , Aging/metabolism , Animals , Calcium/metabolism , Caspase 2/metabolism , Caspase 8/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Ethanol/administration & dosage , Ethanol/pharmacology , Feeding Behavior/drug effects , Fibroblasts/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/enzymology , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...