Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 25(1): 45-67, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38177900

ABSTRACT

Fusion of the outer mitochondrial membrane (OMM) is regulated by mitofusin 1 (MFN1) and 2 (MFN2), yet the differential contribution of each of these proteins is less understood. Mitochondrial carrier homolog 2 (MTCH2) also plays a role in mitochondrial fusion, but its exact function remains unresolved. MTCH2 overexpression enforces MFN2-independent mitochondrial fusion, proposedly by modulating the phospholipid lysophosphatidic acid (LPA), which is synthesized by glycerol-phosphate acyl transferases (GPATs) in the endoplasmic reticulum (ER) and the OMM. Here we report that MTCH2 requires MFN1 to enforce mitochondrial fusion and that fragmentation caused by loss of MTCH2 can be specifically counterbalanced by overexpression of MFN2 but not MFN1, partially independent of its GTPase activity and mitochondrial localization. Pharmacological inhibition of GPATs (GPATi) or silencing ER-resident GPATs suppresses MFN2's ability to compensate for the loss of MTCH2. Loss of either MTCH2, MFN2, or GPATi does not impair stress-induced mitochondrial fusion, whereas the combined loss of MTCH2 and GPATi or the combined loss of MTCH2 and MFN2 does. Taken together, we unmask two cooperative mechanisms that sustain mitochondrial fusion.


Subject(s)
GTP Phosphohydrolases , Lysophospholipids , Mitochondria , Mitochondria/genetics , Mitochondria/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Mitochondrial Dynamics , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
2.
Nat Commun ; 9(1): 5132, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30510213

ABSTRACT

The role of mitochondria dynamics and its molecular regulators remains largely unknown during naïve-to-primed pluripotent cell interconversion. Here we report that mitochondrial MTCH2 is a regulator of mitochondrial fusion, essential for the naïve-to-primed interconversion of murine embryonic stem cells (ESCs). During this interconversion, wild-type ESCs elongate their mitochondria and slightly alter their glutamine utilization. In contrast, MTCH2-/- ESCs fail to elongate their mitochondria and to alter their metabolism, maintaining high levels of histone acetylation and expression of naïve pluripotency markers. Importantly, enforced mitochondria elongation by the pro-fusion protein Mitofusin (MFN) 2 or by a dominant negative form of the pro-fission protein dynamin-related protein (DRP) 1 is sufficient to drive the exit from naïve pluripotency of both MTCH2-/- and wild-type ESCs. Taken together, our data indicate that mitochondria elongation, governed by MTCH2, plays a critical role and constitutes an early driving force in the naïve-to-primed pluripotency interconversion of murine ESCs.


Subject(s)
Mitochondrial Dynamics/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cells, Cultured , Dynamins/genetics , Dynamins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Pluripotent Stem Cells/cytology
3.
Sci Rep ; 7: 44401, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28276496

ABSTRACT

Mitochondrial Carrier Homolog 2 (MTCH2) is a novel regulator of mitochondria metabolism, which was recently associated with Alzheimer's disease. Here we demonstrate that deletion of forebrain MTCH2 increases mitochondria and whole-body energy metabolism, increases locomotor activity, but impairs motor coordination and balance. Importantly, mice deficient in forebrain MTCH2 display a deficit in hippocampus-dependent cognitive functions, including spatial memory, long term potentiation (LTP) and rates of spontaneous excitatory synaptic currents. Moreover, MTCH2-deficient hippocampal neurons display a deficit in mitochondria motility and calcium handling. Thus, MTCH2 is a critical player in neuronal cell biology, controlling mitochondria metabolism, motility and calcium buffering to regulate hippocampal-dependent cognitive functions.


Subject(s)
Calcium/metabolism , Cognition/physiology , Hippocampus/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Neurons/metabolism , Animals , Energy Metabolism/physiology , Female , Hippocampus/physiopathology , Locomotion/physiology , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/deficiency , Neurons/pathology , Postural Balance/physiology , Prosencephalon/metabolism , Prosencephalon/physiopathology , Psychomotor Disorders/metabolism , Psychomotor Disorders/physiopathology , Rotarod Performance Test , Spatial Memory/physiology , Synaptic Transmission/physiology
5.
Obesity (Silver Spring) ; 25(3): 616-625, 2017 03.
Article in English | MEDLINE | ID: mdl-28127879

ABSTRACT

OBJECTIVE: More than one-third of U.S. adults have obesity, causing an alarming increase in obesity-related comorbidities such as type 2 diabetes. The functional role of mitochondrial carrier homolog 2 (MTCH2), a human obesity-associated gene, in lipid homeostasis was investigated in Caenorhabditis elegans, cell culture, and mice. METHODS: In C. elegans, MTCH2/MTCH-1 was depleted, using RNAi and a genetic mutant, and overexpressed to assess its effect on lipid accumulation. In cells and mice, shRNAs against MTCH2 were used for knockdown and MTCH2 overexpression vectors were used for overexpression to study the role of this gene in fat accumulation. RESULTS: MTCH2 knockdown reduced lipid accumulation in adipocyte-like cells in vitro and in C. elegans and mice in vivo. MTCH2 overexpression increased fat accumulation in cell culture, C. elegans, and mice. Acute MTCH2 inhibition reduced fat accumulation in animals subjected to a high-fat diet. Finally, MTCH2 influenced estrogen receptor 1 (ESR1) activity. CONCLUSIONS: MTCH2 is a conserved regulator of lipid homeostasis. MTCH2 was found to be both required and sufficient for lipid homeostasis shifts, suggesting that pharmacological inhibition of MTCH2 could be therapeutic for treatment of obesity and related disorders. MTCH2 could influence lipid homeostasis through inhibition of ESR1 activity.


Subject(s)
Adipocytes/metabolism , Homeostasis/genetics , Lipid Metabolism/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Caenorhabditis elegans , Carrier Proteins/metabolism , Diabetes Mellitus, Type 2 , Diet, High-Fat , Estrogen Receptor alpha/metabolism , Mice , Mitochondrial Membrane Transport Proteins/genetics , Obesity/genetics
6.
Cell Rep ; 14(7): 1602-1610, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26876167

ABSTRACT

Mitochondrial carrier homolog 2 (MTCH2) is a repressor of mitochondrial oxidative phosphorylation (OXPHOS), and its locus is associated with increased BMI in humans. Here, we demonstrate that mice deficient in muscle MTCH2 are protected from diet-induced obesity and hyperinsulinemia and that they demonstrate increased energy expenditure. Deletion of muscle MTCH2 also increases mitochondrial OXPHOS and mass, triggers conversion from glycolytic to oxidative fibers, increases capacity for endurance exercise, and increases heart function. Moreover, metabolic profiling of mice deficient in muscle MTCH2 reveals a preference for carbohydrate utilization and an increase in mitochondria and glycolytic flux in muscles. Thus, MTCH2 is a critical player in muscle biology, modulating metabolism and mitochondria mass as well as impacting whole-body energy homeostasis.


Subject(s)
Metabolome/genetics , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Muscle, Skeletal/metabolism , Obesity/genetics , Animals , Body Composition , Diet, High-Fat , Disease Models, Animal , Energy Metabolism , Gene Expression , Glycolysis/genetics , Humans , Male , Mice , Mice, Knockout , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/deficiency , Muscle, Skeletal/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Oxidative Phosphorylation , Physical Conditioning, Animal
7.
Nat Commun ; 6: 7901, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26219591

ABSTRACT

The metabolic state of stem cells is emerging as an important determinant of their fate. In the bone marrow, haematopoietic stem cell (HSC) entry into cycle, triggered by an increase in intracellular reactive oxygen species (ROS), corresponds to a critical metabolic switch from glycolysis to mitochondrial oxidative phosphorylation (OXPHOS). Here we show that loss of mitochondrial carrier homologue 2 (MTCH2) increases mitochondrial OXPHOS, triggering HSC and progenitor entry into cycle. Elevated OXPHOS is accompanied by an increase in mitochondrial size, increase in ATP and ROS levels, and protection from irradiation-induced apoptosis. In contrast, a phosphorylation-deficient mutant of BID, MTCH2's ligand, induces a similar increase in OXPHOS, but with higher ROS and reduced ATP levels, and is associated with hypersensitivity to irradiation. Thus, our results demonstrate that MTCH2 is a negative regulator of mitochondrial OXPHOS downstream of BID, indispensible in maintaining HSC homeostasis.


Subject(s)
Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , Glycolysis/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Oxidative Phosphorylation , Radiation Tolerance/genetics , Adenosine Triphosphate/metabolism , Animals , Apoptosis/radiation effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Blotting, Western , Cell Cycle/genetics , Cell Differentiation/genetics , Colony-Forming Units Assay , Flow Cytometry , Hematopoietic Stem Cells/cytology , Membrane Potential, Mitochondrial , Mice , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Size , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
8.
J Biol Chem ; 288(30): 22111-27, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23744079

ABSTRACT

Bid is a Bcl-2 family protein that promotes apoptosis by activating Bax and eliciting mitochondrial outer membrane permeabilization (MOMP). Full-length Bid is cleaved in response to apoptotic stimuli into two fragments, p7 and tBid (p15), that are held together by strong hydrophobic interactions until the complex binds to membranes. The detailed mechanism(s) of fragment separation including tBid binding to membranes and release of the p7 fragment to the cytoplasm remain unclear. Using liposomes or isolated mitochondria with fluorescently labeled proteins at physiological concentrations as in vitro models, we report that the two components of the complex quickly separate upon interaction with a membrane. Once tBid binds to the membrane, it undergoes slow structural rearrangements that result in an equilibrium between two major tBid conformations on the membrane. The conformational change of tBid is a prerequisite for interaction with Bax and is, therefore, a novel step that can be modulated to promote or inhibit MOMP. Using automated high-throughput image analysis in cells, we show that down-regulation of Mtch2 causes a significant delay between tBid and Bax relocalization in cells. We propose that by promoting insertion of tBid via a conformational change at the mitochondrial outer membrane, Mtch2 accelerates tBid-mediated Bax activation and MOMP. Thus the interaction of Mtch2 and tBid is a potential target for therapeutic control of Bid initiated cell death.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Membrane/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/chemistry , BH3 Interacting Domain Death Agonist Protein/genetics , Caspase 8/metabolism , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Kinetics , Liposomes/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membranes/physiology , Models, Biological , Models, Molecular , Mutation , Permeability , Protein Binding , Protein Conformation , Time Factors , bcl-2-Associated X Protein/chemistry , bcl-2-Associated X Protein/genetics
9.
Nat Cell Biol ; 14(5): 535-41, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22446738

ABSTRACT

BID, a BH3-only BCL2 family member, functions in apoptosis as well as the DNA-damage response. Our previous data demonstrated that BID is an ATM effector acting to induce cell-cycle arrest and inhibition of apoptosis following DNA damage. Here we show that ATM-mediated BID phosphorylation plays an unexpected role in maintaining the quiescence of haematopoietic stem cells (HSCs). Loss of BID phosphorylation leads to escape from quiescence of HSCs, resulting in exhaustion of the HSC pool and a marked reduction of HSC repopulating potential in vivo. We also demonstrate that BID phosphorylation plays a role in protecting HSCs from irradiation, and that regulating both quiescence and survival of HSCs depends on BID's ability to regulate oxidative stress. Moreover, loss of BID phosphorylation, ATM knockout or exposing mice to irradiation leads to an increase in mitochondrial BID, which correlates with an increase in mitochondrial oxidative stress. These results show that the ATM-BID pathway serves as a critical checkpoint for coupling HSC homeostasis and the DNA-damage stress response to enable long-term regenerative capacity.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/physiology , Cell Cycle Proteins/physiology , Cell Survival/physiology , DNA-Binding Proteins/physiology , Hematopoietic Stem Cells/cytology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , BH3 Interacting Domain Death Agonist Protein/genetics , Cell Cycle Proteins/genetics , DNA Damage , DNA-Binding Proteins/genetics , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics
10.
Nat Cell Biol ; 12(6): 553-562, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20436477

ABSTRACT

The BH3-only BID protein (BH3-interacting domain death agonist) has a critical function in the death-receptor pathway in the liver by triggering mitochondrial outer membrane permeabilization (MOMP). Here we show that MTCH2/MIMP (mitochondrial carrier homologue 2/Met-induced mitochondrial protein), a novel truncated BID (tBID)-interacting protein, is a surface-exposed outer mitochondrial membrane protein that facilitates the recruitment of tBID to mitochondria. Knockout of MTCH2/MIMP in embryonic stem cells and in mouse embryonic fibroblasts hinders the recruitment of tBID to mitochondria, the activation of Bax/Bak, MOMP, and apoptosis. Moreover, conditional knockout of MTCH2/MIMP in the liver decreases the sensitivity of mice to Fas-induced hepatocellular apoptosis and prevents the recruitment of tBID to liver mitochondria both in vivo and in vitro. In contrast, MTCH2/MIMP deletion had no effect on apoptosis induced by other pro-apoptotic Bcl-2 family members and no detectable effect on the outer membrane lipid composition. These loss-of-function models indicate that MTCH2/MIMP has a critical function in liver apoptosis by regulating the recruitment of tBID to mitochondria.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/metabolism , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/physiology , Fibroblasts/metabolism , Mice , Mice, Knockout , Mitochondria, Liver/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Membranes/metabolism , Receptors, Death Domain/metabolism
11.
Cell ; 122(4): 593-603, 2005 Aug 26.
Article in English | MEDLINE | ID: mdl-16122426

ABSTRACT

The "BH3-only" proapoptotic BCL-2 family members are sentinels of intracellular damage. Here, we demonstrated that the BH3-only BID protein partially localizes to the nucleus in healthy cells, is important for apoptosis induced by DNA damage, and is phosphorylated following induction of double-strand breaks in DNA. We also found that BID phosphorylation is mediated by the ATM kinase and occurs in mouse BID on two ATM consensus sites. Interestingly, BID-/- cells failed to accumulate in the S phase of the cell cycle following treatment with the topoisomerase II poison etoposide; reintroducing wild-type BID restored accumulation. In contrast, introducing a nonphosphorylatable BID mutant did not restore accumulation in the S phase and resulted in an increase in cellular sensitivity to etoposide-induced apoptosis. These results implicate BID as an ATM effector and raise the possibility that proapoptotic BID may also play a prosurvival role important for S phase arrest.


Subject(s)
Apoptosis/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , BH3 Interacting Domain Death Agonist Protein , Binding Sites/physiology , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Survival/physiology , Cells, Cultured , DNA/genetics , DNA/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , Etoposide/pharmacology , Fibroblasts/metabolism , Genes, cdc/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/physiology , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , S Phase/drug effects , S Phase/physiology , Topoisomerase II Inhibitors , Tumor Suppressor Proteins/genetics
12.
Mol Cell Biol ; 25(11): 4579-90, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15899861

ABSTRACT

BID, a proapoptotic BCL-2 family member, plays an essential role in the tumor necrosis factor alpha (TNF-alpha)/Fas death receptor pathway in vivo. Activation of the TNF-R1 receptor results in the cleavage of BID into truncated BID (tBID), which translocates to the mitochondria and induces the activation of BAX or BAK. In TNF-alpha-activated FL5.12 cells, tBID becomes part of a 45-kDa cross-linkable mitochondrial complex. Here we describe the biochemical purification of this complex and the identification of mitochondrial carrier homolog 2 (Mtch2) as part of this complex. Mtch2 is a conserved protein that is similar to members of the mitochondrial carrier protein family. Our studies with mouse liver mitochondria indicate that Mtch2 is an integral membrane protein exposed on the surface of mitochondria. Using blue-native gel electrophoresis we revealed that in viable FL5.12 cells Mtch2 resides in a protein complex of ca. 185 kDa and that the addition of TNF-alpha to these cells leads to the recruitment of tBID and BAX to this complex. Importantly, this recruitment was partially inhibited in FL5.12 cells stably expressing BCL-X(L). These results implicate Mtch2 as a mitochondrial target of tBID and raise the possibility that the Mtch2-resident complex participates in the mitochondrial apoptotic program.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Sequence , Animals , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/analysis , Conserved Sequence , Humans , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Membrane Transport Proteins/analysis , Membrane Transport Proteins/genetics , Mice , Mitochondria/chemistry , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/analysis , Mitochondrial Proteins/genetics , Molecular Sequence Data , Proto-Oncogene Proteins c-bcl-2/physiology , Signal Transduction , bcl-X Protein
13.
J Biol Chem ; 278(12): 10707-15, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12519725

ABSTRACT

The proapoptotic activity of BID seems to solely depend upon its cleavage to truncated tBID. Here we demonstrate that expression of a caspase-8 non-cleavable (nc) BID-D59A mutant or expression of wild type (wt) BID induces apoptosis in Bid -/-, caspase-8 -/-, and wt primary MEFs. Western blot analysis indicated that no cleavage products appeared in cells expressing ncBID. ncBID was as effective as wtBID in inducing cytochrome c release, caspase activation, and apoptosis. ncBID and wtBID (nc/wtBID) were much less effective than tBID in localizing to mitochondria and in inducing cytochrome c release, but only slightly less effective in inducing apoptosis. Studies with Apaf-1- and caspase-9-deficient primary MEFs indicated that both proteins were essential for nc/wtBID and for tBID-induced apoptosis. Most importantly, expression of non-apoptotic levels of either ncBID or wtBID in Bid -/- MEFs induced a similar and significant enhancement in apoptosis in response to a variety of death signals, which was accompanied by enhanced localization of BID to mitochondria and cytochrome c release. Thus, these results implicate full-length BID as an active player in the mitochondria during apoptosis.


Subject(s)
Apoptosis , Carrier Proteins/physiology , Animals , Apoptotic Protease-Activating Factor 1 , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/chemistry , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/physiology , Cells, Cultured , Cytochrome c Group/metabolism , Mice , Mitochondria/enzymology , Mutation , Proteins/physiology , Simian virus 40/genetics
14.
J Biol Chem ; 277(14): 12237-45, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11805084

ABSTRACT

Activation of the tumor necrosis factor R1/Fas receptor results in the cleavage of cytosolic BID to truncated tBID. tBID translocates to the mitochondria to induce the oligomerization of BAX or BAK, resulting in the release of cytochrome c (Cyt c). Here we demonstrate that in tumor necrosis factor alpha-activated FL5.12 cells, tBID becomes part of a 45-kDa cross-linkable mitochondrial complex that does not include BAX or BAK. Using fluorescence resonance energy transfer analysis and co-immunoprecipitation, we demonstrate that tBID-tBID interactions occur in the mitochondria of living cells. Cross-linking experiments using a tBID-GST chimera indicated that tBID forms homotrimers in the mitochondrial membrane. To test the functional consequence of tBID oligomerization, we expressed a chimeric FKBP-tBID molecule. Enforced dimerization of FKBP-tBID by the bivalent ligand FK1012 resulted in Cyt c release, caspase activation, and apoptosis. Surprisingly, enforced dimerization of tBID did not result in the dimerization of either BAX or BAK. Moreover, a tBID BH3 mutant (G94E), which does not interact with or induce the dimerization of either BAX or BAK, formed the 45-kDa complex and induced both Cyt c release and apoptosis. Thus, tBID oligomerization may represent an alternative mechanism for inducing mitochondrial dysfunction and apoptosis.


Subject(s)
Apoptosis , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Intracellular Membranes/metabolism , Mitochondria/metabolism , Animals , BH3 Interacting Domain Death Agonist Protein , Blotting, Western , COS Cells , Caspase 3 , Caspases/metabolism , Cell Line , Cross-Linking Reagents/pharmacology , Cytosol/metabolism , Dimerization , HeLa Cells , Humans , Ligands , Mice , Microscopy, Confocal , Models, Biological , Plasmids/metabolism , Precipitin Tests , Protein Binding , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Subcellular Fractions , Time Factors , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...