Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 4583, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321484

ABSTRACT

Voltage dependent anion channel 2 (VDAC2) is an outer mitochondrial membrane porin known to play a significant role in apoptosis and calcium signaling. Abnormalities in calcium homeostasis often leads to electrical and contractile dysfunction and can cause dilated cardiomyopathy and heart failure. However, the specific role of VDAC2 in intracellular calcium dynamics and cardiac function is not well understood. To elucidate the role of VDAC2 in calcium homeostasis, we generated a cardiac ventricular myocyte-specific developmental deletion of Vdac2 in mice. Our results indicate that loss of VDAC2 in the myocardium causes severe impairment in excitation-contraction coupling by altering both intracellular and mitochondrial calcium signaling. We also observed adverse cardiac remodeling which progressed to severe cardiomyopathy and death. Reintroduction of VDAC2 in 6-week-old knock-out mice partially rescued the cardiomyopathy phenotype. Activation of VDAC2 by efsevin increased cardiac contractile force in a mouse model of pressure-overload induced heart failure. In conclusion, our findings demonstrate that VDAC2 plays a crucial role in cardiac function by influencing cellular calcium signaling. Through this unique role in cellular calcium dynamics and excitation-contraction coupling VDAC2 emerges as a plausible therapeutic target for heart failure.


Subject(s)
Calcium/metabolism , Cardiomyopathy, Dilated/metabolism , Homeostasis , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channel 2/metabolism , Animals , Apoptosis , Calcium Signaling , Cardiomyopathy, Dilated/mortality , Heart Failure/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Transcriptome
2.
Elife ; 2: e00772, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23991283

ABSTRACT

A critical event in ischemia-based cell death is the opening of the mitochondrial permeability transition pore (MPTP). However, the molecular identity of the components of the MPTP remains unknown. Here, we determined that the Bcl-2 family members Bax and Bak, which are central regulators of apoptotic cell death, are also required for mitochondrial pore-dependent necrotic cell death by facilitating outer membrane permeability of the MPTP. Loss of Bax/Bak reduced outer mitochondrial membrane permeability and conductance without altering inner membrane MPTP function, resulting in resistance to mitochondrial calcium overload and necrotic cell death. Reconstitution with mutants of Bax that cannot oligomerize and form apoptotic pores, but still enhance outer membrane permeability, permitted MPTP-dependent mitochondrial swelling and restored necrotic cell death. Our data predict that the MPTP is an inner membrane regulated process, although in the absence of Bax/Bak the outer membrane resists swelling and prevents organelle rupture to prevent cell death. DOI:http://dx.doi.org/10.7554/eLife.00772.001.


Subject(s)
Mitochondrial Membrane Transport Proteins/physiology , Necrosis , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology , Animals , Mice , Mitochondrial Permeability Transition Pore
3.
Sci Signal ; 6(268): ra20, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23532334

ABSTRACT

The clinical efficacy of tyrosine kinase inhibitors supports the dependence of distinct subsets of cancers on specific driver mutations for survival, a phenomenon called "oncogene addiction." We demonstrate that PUMA and BIM are the key apoptotic effectors of tyrosine kinase inhibitors in breast cancers with amplification of the gene encoding human epidermal growth factor receptor 2 (HER2) and lung cancers with epidermal growth factor receptor (EGFR) mutants. The BH3 domain containing proteins BIM and PUMA can directly activate the proapoptotic proteins BAX and BAK to permeabilize mitochondria, leading to caspase activation and apoptosis. We delineated the signal transduction pathways leading to the induction of BIM and PUMA by tyrosine kinase inhibitors. Inhibition of the mitogen-activated or extracellular signal-regulated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway caused increased abundance of BIM, whereas antagonizing the phosphoinositide 3-kinase (PI3K)-AKT pathway triggered nuclear translocation of the FOXO transcription factors, which directly activated the PUMA promoter. In a mouse breast tumor model, the abundance of PUMA and BIM was increased after inactivation of HER2. Moreover, deficiency of Bim or Puma impaired caspase activation and reduced tumor regression caused by inactivation of HER2. Similarly, deficiency of Puma impeded the regression of EGFR(L858R)-driven mouse lung tumors upon inactivation of the EGFR-activating mutant. Overall, our study identified PUMA and BIM as the sentinels that interconnect kinase signaling networks and the mitochondrion-dependent apoptotic program, which offers therapeutic insights for designing novel cell death mechanism-based anticancer strategies.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/genetics , Breast Neoplasms/metabolism , Gene Silencing/physiology , Membrane Proteins/metabolism , Oncogenes/physiology , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Biphenyl Compounds/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Chromatin Immunoprecipitation , ErbB Receptors/metabolism , Female , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Lapatinib , MAP Kinase Signaling System/physiology , Membrane Proteins/genetics , Mice , Nitrophenols/pharmacology , Oncogenes/genetics , Piperazines/pharmacology , Plasmids/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Quinazolines , RNA Interference , RNA, Small Interfering/genetics , Receptor, ErbB-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Sulfonamides/pharmacology
4.
Cancer Res ; 72(3): 736-46, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22166309

ABSTRACT

The threonine endopeptidase Taspase1 has a critical role in cancer cell proliferation and apoptosis. In this study, we developed and evaluated small molecule inhibitors of Taspase1 as a new candidate class of therapeutic modalities. Genetic deletion of Taspase1 in the mouse produced no overt deficiencies, suggesting the possibility of a wide therapeutic index for use of Taspase1 inhibitors in cancers. We defined the peptidyl motifs recognized by Taspase1 and conducted a cell-based dual-fluorescent proteolytic screen of the National Cancer Institute diversity library to identify Taspase1 inhibitors (TASPIN). On the basis of secondary and tertiary screens the 4-[(4-arsonophenyl)methyl]phenyl] arsonic acid NSC48300 was determined to be the most specific active compound. Structure-activity relationship studies indicated a crucial role for the arsenic acid moiety in mediating Taspase1 inhibition. Additional fluorescence resonance energy transfer-based kinetic analysis characterized NSC48300 as a reversible, noncompetitive inhibitor of Taspase1 (K(i) = 4.22 µmol/L). In the MMTV-neu mouse model of breast cancer and the U251 xenograft model of brain cancer, NSC48300 produced effective tumor growth inhibition. Our results offer an initial preclinical proof-of-concept to develop TASPINs for cancer therapy.


Subject(s)
Arsenicals/pharmacology , Brain Neoplasms/prevention & control , Breast Neoplasms/prevention & control , Endopeptidases/metabolism , Protease Inhibitors/pharmacology , Amino Acid Sequence , Amino Acids/genetics , Amino Acids/metabolism , Animals , Binding Sites/genetics , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Endopeptidases/genetics , HEK293 Cells , Humans , Kinetics , Male , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Sequence Homology, Amino Acid , Small Molecule Libraries , Xenograft Model Antitumor Assays
5.
EMBO J ; 30(16): 3383-96, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21750527

ABSTRACT

Genomic instability due to telomere dysfunction and defective repair of DNA double-strand breaks (DSBs) is an underlying cause of ageing-related diseases. 53BP1 is a key factor in DNA DSBs repair and its deficiency is associated with genomic instability and cancer progression. Here, we uncover a novel pathway regulating the stability of 53BP1. We demonstrate an unprecedented role for the cysteine protease Cathepsin L (CTSL) in the degradation of 53BP1. Overexpression of CTSL in wild-type fibroblasts leads to decreased 53BP1 protein levels and changes in its cellular distribution, resulting in defective repair of DNA DSBs. Importantly, we show that the defects in DNA repair associated with 53BP1 deficiency upon loss of A-type lamins are due to upregulation of CTSL. Furthermore, we demonstrate that treatment with vitamin D stabilizes 53BP1 and promotes DNA DSBs repair via inhibition of CTSL, providing an as yet unsuspected link between vitamin D action and DNA repair. Given that CTSL upregulation is a hallmark of cancer and progeria, regulation of this pathway could be of great therapeutic significance for these diseases.


Subject(s)
Cathepsin L/physiology , Chromosomal Proteins, Non-Histone/metabolism , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Lamin Type A/physiology , Vitamin D/physiology , Animals , Calcitriol/pharmacology , Cathepsin L/antagonists & inhibitors , Cathepsin L/biosynthesis , Cathepsin L/genetics , Cell Line , Enzyme Precursors/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lamin Type A/deficiency , Lamin Type A/genetics , Leupeptins/pharmacology , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Recombinant Fusion Proteins/physiology , Species Specificity , Transfection , Tumor Suppressor p53-Binding Protein 1
6.
Science ; 330(6009): 1390-3, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21127253

ABSTRACT

Although the proteins BAX and BAK are required for initiation of apoptosis at the mitochondria, how BAX and BAK are activated remains unsettled. We provide in vivo evidence demonstrating an essential role of the proteins BID, BIM, and PUMA in activating BAX and BAK. Bid, Bim, and Puma triple-knockout mice showed the same developmental defects that are associated with deficiency of Bax and Bak, including persistent interdigital webs and imperforate vaginas. Genetic deletion of Bid, Bim, and Puma prevented the homo-oligomerization of BAX and BAK, and thereby cytochrome c-mediated activation of caspases in response to diverse death signals in neurons and T lymphocytes, despite the presence of other BH3-only molecules. Thus, many forms of apoptosis require direct activation of BAX and BAK at the mitochondria by a member of the BID, BIM, or PUMA family of proteins.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Membrane Proteins/metabolism , Neurons/physiology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , BH3 Interacting Domain Death Agonist Protein/deficiency , BH3 Interacting Domain Death Agonist Protein/genetics , Bcl-2-Like Protein 11 , Caspases/metabolism , Cells, Cultured , Cerebellum/cytology , Cytochromes c/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondria/metabolism , Models, Biological , Permeability , Protein Multimerization , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Stress, Physiological , T-Lymphocytes/physiology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , bcl-2 Homologous Antagonist-Killer Protein/chemistry , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/chemistry , bcl-2-Associated X Protein/genetics
7.
Nature ; 467(7313): 343-6, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20818375

ABSTRACT

Cell cycle checkpoints are implemented to safeguard the genome, avoiding the accumulation of genetic errors. Checkpoint loss results in genomic instability and contributes to the evolution of cancer. Among G1-, S-, G2- and M-phase checkpoints, genetic studies indicate the role of an intact S-phase checkpoint in maintaining genome integrity. Although the basic framework of the S-phase checkpoint in multicellular organisms has been outlined, the mechanistic details remain to be elucidated. Human chromosome-11 band-q23 translocations disrupting the MLL gene lead to poor prognostic leukaemias. Here we assign MLL as a novel effector in the mammalian S-phase checkpoint network and identify checkpoint dysfunction as an underlying mechanism of MLL leukaemias. MLL is phosphorylated at serine 516 by ATR in response to genotoxic stress in the S phase, which disrupts its interaction with, and hence its degradation by, the SCF(Skp2) E3 ligase, leading to its accumulation. Stabilized MLL protein accumulates on chromatin, methylates histone H3 lysine 4 at late replication origins and inhibits the loading of CDC45 to delay DNA replication. Cells deficient in MLL showed radioresistant DNA synthesis and chromatid-type genomic abnormalities, indicative of S-phase checkpoint dysfunction. Reconstitution of Mll(-/-) (Mll also known as Mll1) mouse embryonic fibroblasts with wild-type but not S516A or ΔSET mutant MLL rescues the S-phase checkpoint defects. Moreover, murine myeloid progenitor cells carrying an Mll-CBP knock-in allele that mimics human t(11;16) leukaemia show a severe radioresistant DNA synthesis phenotype. MLL fusions function as dominant negative mutants that abrogate the ATR-mediated phosphorylation/stabilization of wild-type MLL on damage to DNA, and thus compromise the S-phase checkpoint. Together, our results identify MLL as a key constituent of the mammalian DNA damage response pathway and show that deregulation of the S-phase checkpoint incurred by MLL translocations probably contributes to the pathogenesis of human MLL leukaemias.


Subject(s)
Cell Cycle Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , S Phase/physiology , Alleles , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Line , Chromatin/metabolism , DNA Damage , DNA Replication/physiology , Genes, Dominant/genetics , Genomic Instability/physiology , Histone-Lysine N-Methyltransferase , Histones/chemistry , Histones/metabolism , Humans , Leukemia/genetics , Lysine/metabolism , Methylation , Mice , Myeloid Progenitor Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/chemistry , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Phosphorylation , Phosphoserine/metabolism , Protein Binding , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Translocation, Genetic/genetics
8.
Cancer Res ; 70(13): 5358-67, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20516119

ABSTRACT

Taspase1, the mixed lineage leukemia and TFIIAalpha-beta cleaving protease, enables cell proliferation and permits oncogenic initiation. Here, we show its critical role in cancer maintenance and thus offer a new anticancer target. Taspase1 is overexpressed in primary human cancers, and deficiency of Taspase1 in cancer cells not only disrupts proliferation but also enhances apoptosis. Mechanistically, loss of Taspase1 induces the levels of CDK inhibitors (CDKI: p16, p21, and p27) and reduces the level of antiapoptotic MCL-1. Therapeutically, deficiency of Taspase1 synergizes with chemotherapeutic agents and ABT-737, an inhibitor of BCL-2/BCL-X(L), to kill cancer cells. Taspase1 alone or in conjunction with MYC, RAS, or E1A fails to transform NIH/3T3 cells or primary mouse embryonic fibroblasts, respectively, but plays critical roles in cancer initiation and maintenance. Therefore, Taspase1 is better classified as a "non-oncogene addiction" protease, the inhibition of which may offer a novel anticancer therapeutic strategy. The reliance of oncogenes on subordinate non-oncogenes during tumorigenesis underscores the non-oncogene addiction hypothesis in which a large class of non-oncogenes functions to maintain cancer phenotypes and presents attractive anticancer therapeutic targets. The emergence of successful cancer therapeutics targeting non-oncogenes to which cancers are addicted supports the future development and potential application of small-molecule Taspase1 inhibitors for cancer therapy.


Subject(s)
Endopeptidases/genetics , Glioblastoma/genetics , Melanoma/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Biphenyl Compounds/pharmacology , Cell Growth Processes/genetics , Cell Line, Transformed , Cell Line, Tumor , Endopeptidases/deficiency , Endopeptidases/metabolism , Genes, myc , Genes, ras , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein , NIH 3T3 Cells , Nitrophenols/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , Transduction, Genetic , Transfection
9.
Proc Natl Acad Sci U S A ; 107(20): 9035-42, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20418503

ABSTRACT

Dysregulation of programmed cell death due to abnormal expression of Bcl-2 proteins is implicated in cancer, neurodegenerative diseases, and heart failure. Among Bcl-2 family members, BNip proteins uniquely stimulate cell death with features of both apoptosis and necrosis. Localization of these factors to mitochondria and endoplasmic reticulum (ER) provides additional complexity. Previously, we observed regulation of intracellular calcium stores by reticular Nix. Here, we report effects of Nix targeting to mitochondria or ER on cell death pathways and heart failure progression. Nix-deficient fibroblasts expressing mitochondrial-directed or ER-directed Nix mutants exhibited similar cytochrome c release, caspase activation, annexin V and TUNEL labeling, and cell death. ER-Nix cells, but not mitochondrial-Nix cells, showed dissipation of mitochondrial inner membrane potential, Deltapsi(m), and were protected from cell death by cyclosporine A or ppif ablation, implicating the mitochondrial permeability transition pore (MPTP). ER-Nix cells were not protected from death by caspase inhibition or combined ablation of Bax and Bak. Combined inhibition of caspases and the MPTP fully protected against Nix-mediated cell death. To determine the role of the dual pathways in heart failure, mice conditionally overexpressing Nix or Nix mutants in hearts were created. Cardiomyocte death caused by mitochondrial- and ER-directed Nix was equivalent, but ppif ablation fully protected only ER-Nix. Thus, Nix stimulates dual autonomous death pathways, determined by its subcellular localization. Mitochondrial Nix activates Bax/Bak- and caspase-dependent apoptosis, whereas ER-Nix activates Bax/Bak-independent, MPTP-dependent necrosis. Complete protection against programmed cell death mediated by Nix and related factors can be achieved by simultaneous inhibition of both pathways.


Subject(s)
Apoptosis/physiology , Cardiomyopathies/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Necrosis/metabolism , Signal Transduction/physiology , Animals , Caspases/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , In Situ Nick-End Labeling , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Mutant Strains , Microscopy, Fluorescence , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
10.
Mol Cell ; 36(3): 487-99, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19917256

ABSTRACT

While activation of BAX/BAK by BH3-only molecules (BH3s) is essential for mitochondrial apoptosis, the underlying mechanisms remain unsettled. Here we demonstrate that BAX undergoes stepwise structural reorganization leading to mitochondrial targeting and homo-oligomerization. The alpha1 helix of BAX keeps the alpha9 helix engaged in the dimerization pocket, rendering BAX as a monomer in cytosol. The activator BH3s, tBID/BIM/PUMA, attack and expose the alpha1 helix of BAX, resulting in secondary disengagement of the alpha9 helix and thereby mitochondrial insertion. Activator BH3s remain associated with the N-terminally exposed BAX through the BH1 domain to drive homo-oligomerization. BAK, an integral mitochondrial membrane protein, has bypassed the first activation step, explaining why its killing kinetics are faster than those of BAX. Furthermore, death signals initiated at ER induce BIM and PUMA to activate mitochondrial apoptosis. Accordingly, deficiency of Bim/Puma impedes ER stress-induced BAX/BAK activation and apoptosis. Our study provides mechanistic insights regarding the spatiotemporal execution of BAX/BAK-governed cell death.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , Bcl-2-Like Protein 11 , Cells, Cultured , Etoposide/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Immunoprecipitation , Membrane Proteins/genetics , Mice , Mice, Knockout , Microscopy, Fluorescence , Mitochondria/metabolism , Models, Biological , Mutation , Protein Binding/drug effects , Protein Multimerization , Proto-Oncogene Proteins/genetics , Staurosporine/pharmacology , Thapsigargin/pharmacology , Tumor Suppressor Proteins/genetics , Tunicamycin/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/chemistry , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/chemistry , bcl-2-Associated X Protein/genetics
11.
Cancer Biol Ther ; 8(13): 1204-11, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19729989

ABSTRACT

Human leukemias with chromosomal band 11q23 aberrations that disrupt the MLL/HRX/ALL-1 gene portend poor prognosis. MLL associated leukemias account for the majority of infant leukemia, approximately 10% of adult de novo leukemia and approximately 33% of therapy related acute leukemia with a balanced chromosome translocation. The 500 kD MLL precursor is processed by Taspase1 to generate mature MLL(N320/C180), which orchestrates many aspects of biology such as embryogenesis, cell cycle, cell fate and stem cell maintenance. Leukemogenic MLL translocations fuse the common MLL N-terminus (approximately 1,400 aa) in frame with more than 60 translocation partner genes (TPGs). Recent studies on MLL and MLL leukemia have greatly advanced our knowledge concerning the normal function of MLL and its deregulation in leukemogenesis. Here, we summarize the critical biological and pathological activities of MLL and MLL fusions, and discuss available models and potential therapeutic targets of MLL associated leukemias.


Subject(s)
Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Translocation, Genetic , Animals , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Leukemia/pathology , Models, Biological , Signal Transduction/genetics
12.
Sci Signal ; 2(85): ra48, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19706873

ABSTRACT

The proapoptotic proteins BAX and BAK constitute the mitochondrial apoptotic gateway that executes cellular demise after integrating death signals. The lethal BAK is kept in check by voltage-dependent anion channel 2 (VDAC2), a mammalian-restricted VDAC isoform. Here, we provide evidence showing a critical role for the VADC2-BAK complex in determining thymocyte survival in vivo. Genetic depletion of Vdac2 in the thymus resulted in excessive cell death and hypersensitivity to diverse death stimuli including engagement of the T cell receptor. These phenotypes were completely rescued by the concurrent deletion of Bak but not that of Bax. Thus, the VDAC2-BAK axis provides a mechanism that governs the homeostasis of thymocytes. Our study reveals a sophisticated built-in rheostat that likely fine-tunes immune competence to balance autoimmunity and immunodeficiency.


Subject(s)
Clonal Deletion/physiology , T-Lymphocytes/cytology , Voltage-Dependent Anion Channel 2/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/genetics , Apoptosis/physiology , Autoimmunity/genetics , Autoimmunity/physiology , CD3 Complex/immunology , Clonal Deletion/genetics , Dimerization , Female , Gene Knockout Techniques , Genotype , Ion Transport/genetics , Ion Transport/physiology , Male , Mice , Mice, Knockout , Mitochondrial Membranes/physiology , Thymus Gland/cytology , Voltage-Dependent Anion Channel 2/deficiency , Voltage-Dependent Anion Channel 2/genetics , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/physiology
13.
Mol Cell ; 33(2): 145-6, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19187757

ABSTRACT

Activation of Bax and Bak by BH3-only molecules triggers mitochondrial apoptosis. In a recent issue of Molecular Cell, Fu et al. (2009) identify a constitutively active isoform of Bax, Baxbeta, whose activity is tightly controlled by the ubiquitin-proteasome system.


Subject(s)
Alternative Splicing/physiology , bcl-2-Associated X Protein/metabolism , Apoptosis , Mitochondria/metabolism , Models, Biological , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms/metabolism , Ubiquitins/metabolism
14.
Proc Natl Acad Sci U S A ; 106(4): 1093-8, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19144918

ABSTRACT

Three forms of cell death have been described: apoptosis, autophagic cell death, and necrosis. Although genetic and biochemical studies have formulated a detailed blueprint concerning the apoptotic network, necrosis is generally perceived as a passive cellular demise resulted from unmanageable physical damages. Here, we conclude an active de novo genetic program underlying DNA damage-induced necrosis, thus assigning necrotic cell death as a form of "programmed cell death." Cells deficient of the essential mitochondrial apoptotic effectors, BAX and BAK, ultimately succumbed to DNA damage, exhibiting signature necrotic characteristics. Importantly, this genotoxic stress-triggered necrosis was abrogated when either transcription or translation was inhibited. We pinpointed the p53-cathepsin axis as the quintessential framework underlying necrotic cell death. p53 induces cathepsin Q that cooperates with reactive oxygen species (ROS) to execute necrosis. Moreover, we presented the in vivo evidence of p53-activated necrosis in tumor allografts. Current study lays the foundation for future experimental and therapeutic discoveries aimed at "programmed necrotic death."


Subject(s)
Cathepsins/metabolism , Cysteine Endopeptidases/metabolism , DNA Damage , Necrosis/pathology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cathepsins/genetics , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Fibroblasts/pathology , Fibroblasts/ultrastructure , Humans , Mice , Neoplasm Transplantation , Neoplasms/pathology , Neoplasms/ultrastructure , Transcriptional Activation/genetics , Transplantation, Homologous , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/metabolism
15.
Nature ; 455(7216): 1076-81, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18948948

ABSTRACT

BAX is a pro-apoptotic protein of the BCL-2 family that is stationed in the cytosol until activated by a diversity of stress stimuli to induce cell death. Anti-apoptotic proteins such as BCL-2 counteract BAX-mediated cell death. Although an interaction site that confers survival functionality has been defined for anti-apoptotic proteins, an activation site has not been identified for BAX, rendering its explicit trigger mechanism unknown. We previously developed stabilized alpha-helix of BCL-2 domains (SAHBs) that directly initiate BAX-mediated mitochondrial apoptosis. Here we demonstrate by NMR analysis that BIM SAHB binds BAX at an interaction site that is distinct from the canonical binding groove characterized for anti-apoptotic proteins. The specificity of the human BIM-SAHB-BAX interaction is highlighted by point mutagenesis that disrupts functional activity, confirming that BAX activation is initiated at this novel structural location. Thus, we have now defined a BAX interaction site for direct activation, establishing a new target for therapeutic modulation of apoptosis.


Subject(s)
Gene Expression Regulation , bcl-2-Associated X Protein/metabolism , Amino Acid Sequence , Animals , Apoptosis , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Bcl-2-Like Protein 11 , Cell Line , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mutagenesis, Site-Directed , Mutation/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Sequence Alignment , bcl-2-Associated X Protein/chemistry
16.
Cell Cycle ; 7(4): 428-35, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18235233

ABSTRACT

Discovered in 1992 from cloning the gene involved in human leukemias carrying chromosome band 11q23 translocations, the MLL/HRX/ALL-1 gene has since attracted scientists from various disciplines by its diverse functions in normal physiological and pathological processes. MLL is the human orthologue of Drosophila trithorax (trx)-the founding member of trithorax group proteins, Trx-G. Leukemogenic11q23 translocations fuse the common MLL N-terminal 1400aa in-frame with a wide variety of fusion partners that share no structural or functional homology. The 500 kD precursor MLL undergoes evolutionarily conserved site-specific cleavage mediated by Taspase1, generating the mature MLL(N320/C180) heterodimer which methylates histone H3 at lysine 4 with its carboxy-terminal SET domain. Extensive biochemical and genetic studies on MLL/trx have established its critical role in maintaining the expression of Hox/homeotic genes. By contrast, the involvement of MLL in many other essential cellular processes remains unclear. Recent reports including ours began to elucidate the intricate interplay between MLL and the cell cycle machinery, which ensures proper cell cycle phase transitions. Thus, this review will focus on this novel activity of MLL and discuss the implications of its deregulation in MLL leukemias.


Subject(s)
Cell Cycle/physiology , Endopeptidases/metabolism , Gene Expression Regulation, Neoplastic/physiology , Leukemia/metabolism , Models, Biological , Myeloid-Lymphoid Leukemia Protein/metabolism , Signal Transduction/physiology , Humans , Myeloid-Lymphoid Leukemia Protein/biosynthesis
17.
Int J Radiat Oncol Biol Phys ; 70(3): 804-15, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18191336

ABSTRACT

PURPOSE: To address in vivo the issue of whether Bax and Bak are functionally redundant in signaling apoptosis, capable of substituting for each other. METHODS AND MATERIALS: Mice were exposed to whole-body radiation, and endothelial cell apoptosis was quantified using double immunostaining with TUNEL and anti-CD31 antibody. Crypt survival was determined at 3.5 days after whole-body radiation by the microcolony survival assay. Actuarial animal survival was calculated by the product-limit Kaplan-Meier method, and autopsies were performed to establish cause of death. RESULTS: Radiation exposure of Bax- and Bak-deficient mice, both expressing a wild-type acid sphingomyelinase (ASMase) phenotype, indicated that Bax and Bak are both mandatory, though mutually independent, for the intestinal endothelial apoptotic response. However, neither affected epithelial apoptosis at crypt positions 4-5, indicating specificity toward endothelium. Furthermore, Bax deficiency and Bak deficiency each individually mimicked ASMase deficiency in inhibiting crypt lethality in the microcolony assay and in rescuing mice from the lethal gastrointestinal syndrome. CONCLUSIONS: The data indicate that Bax and Bak have nonredundant functional roles in the apoptotic response of the irradiated intestinal endothelium. The observation that Bax deficiency and Bak deficiency also protect crypts in the microcolony assay provides strong evidence that the microvascular apoptotic component is germane to the mechanism of radiation-induced damage to mouse intestines, regulating reproductive cell death of crypt stem cell clonogens.


Subject(s)
Apoptosis , Endothelial Cells/radiation effects , Intestinal Mucosa/radiation effects , Intestine, Small/radiation effects , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology , Animals , Biomarkers/metabolism , Colony-Forming Units Assay , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/radiation effects , Female , In Situ Nick-End Labeling , Intestinal Mucosa/cytology , Intestine, Small/cytology , Jejunum/cytology , Jejunum/radiation effects , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Radiation Injuries/etiology , Radiation Injuries/pathology , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism , Whole-Body Irradiation , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2-Associated X Protein/deficiency
18.
Genes Dev ; 21(19): 2385-98, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17908926

ABSTRACT

Human chromosome 11q23 translocations disrupting MLL result in poor prognostic leukemias. It fuses the common MLL N-terminal approximately 1400 amino acids in-frame with >60 different partners without shared characteristics. In addition to the well-characterized activity of MLL in maintaining Hox gene expression, our recent studies established an MLL-E2F axis in orchestrating core cell cycle gene expression including Cyclins. Here, we demonstrate a biphasic expression of MLL conferred by defined windows of degradation mediated by specialized cell cycle E3 ligases. Specifically, SCF(Skp2) and APC(Cdc20) mark MLL for degradation at S phase and late M phase, respectively. Abolished peak expression of MLL incurs corresponding defects in G1/S transition and M-phase progression. Conversely, overexpression of MLL blocks S-phase progression. Remarkably, MLL degradation initiates at its N-terminal approximately 1400 amino acids, and tested prevalent MLL fusions are resistant to degradation. Thus, impaired degradation of MLL fusions likely constitutes the universal mechanism underlying all MLL leukemias. Our data conclude an essential post-translational regulation of MLL by the cell cycle ubiquitin/proteasome system (UPS) assures the temporal necessity of MLL in coordinating cell cycle progression.


Subject(s)
Cell Cycle Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Cdc20 Proteins , Cell Cycle , HeLa Cells , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Myeloid/etiology , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Protein Processing, Post-Translational
19.
Nat Cell Biol ; 8(12): 1348-58, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17115033

ABSTRACT

Although the BCL-2 family constitutes a crucial checkpoint in apoptosis, the intricate interplay between these family members remains elusive. Here, we demonstrate that BIM and PUMA, similar to truncated BID (tBID), directly activate BAX-BAK to release cytochrome c. Conversely, anti-apoptotic BCL-2-BCL-X(L)-MCL-1 sequesters these 'activator' BH3-only molecules into stable complexes, thus preventing the activation of BAX-BAK. Extensive mutagenesis of BAX-BAK indicates that their activity is not kept in check by BCL-2-BCL-X(L)-MCL-1. Anti-apoptotic BCL-2 members are differentially inactivated by the remaining 'inactivator' BH3-only molecules including BAD, NOXA, BMF, BIK/BLK and HRK/DP5. BAD displaces tBID, BIM or PUMA from BCL-2-BCL-X(L) to activate BAX-BAK, whereas NOXA specifically antagonizes MCL-1. Coexpression of BAD and NOXA killed wild-type but not Bax, Bak doubly deficient cells or Puma deficient cells with Bim knockdown, indicating that activator BH3-only molecules function downstream of inactivator BH3-only molecules to activate BAX-BAK. Our data establish a hierarchical regulation of mitochondrion-dependent apoptosis by various BCL-2 subfamilies.


Subject(s)
Apoptosis , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Bcl-2-Like Protein 11 , Cytochromes c/metabolism , Membrane Proteins/metabolism , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
20.
Genes Dev ; 20(17): 2397-409, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951254

ABSTRACT

Taspase1 was identified as the threonine endopeptidase that cleaves mixed-lineage leukemia (MLL) for proper Hox gene expression in vitro. To investigate its functions in vivo, we generated Taspase1(-/-) mice. Taspase1 deficiency results in noncleavage (nc) of MLL and MLL2 and homeotic transformations. Remarkably, our in vivo studies uncover an unexpected role of Taspase1 in the cell cycle. Taspase1(-/-) animals are smaller in size. Taspase1(-/-) mouse embryonic fibroblasts (MEFs) exhibit impaired proliferation, and acute deletion of Taspase1 leads to a marked reduction of thymocytes. Taspase1 deficiency incurs down-regulation of Cyclin Es, As, and Bs and up-regulation of p16(Ink4a) . We show that MLL and MLL2 directly target E2Fs for Cyclin expression. The uncleaved precursor MLL displays a reduced histone H3 methyl transferase activity in vitro. Accordingly, chromatin immunoprecipitation assays demonstrate a markedly decreased histone H3 K4 trimethylation at Cyclin E1 and E2 genes in Taspase1(-/-) cells. Furthermore, MLL(nc/nc;2nc/nc) MEFs are also impaired in proliferation. Our data are consistent with a model in which precursor MLLs, activated by Taspase1, target to Cyclins through E2Fs to methylate histone H3 at K4, leading to activation. Lastly, Taspase1(-/-) cells are resistant to oncogenic transformation, and Taspase1 is overexpressed in many cancer cell lines. Thus, Taspase1 may serve as a target for cancer therapeutics.


Subject(s)
Cell Cycle/physiology , Endopeptidases/physiology , Myeloid-Lymphoid Leukemia Protein/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Endopeptidases/biosynthesis , Endopeptidases/deficiency , Endopeptidases/genetics , Hydrolysis , Mice , Mice, Knockout , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL