Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Blood Cancer J ; 14(1): 16, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38253636

ABSTRACT

Plk1-interacting checkpoint helicase (PICH) is a DNA translocase involved in resolving ultrafine anaphase DNA bridges and, therefore, is important to safeguard chromosome segregation and stability. PICH is overexpressed in various human cancers, particularly in lymphomas such as Burkitt lymphoma, which is caused by MYC translocations. To investigate the relevance of PICH in cancer development and progression, we have combined novel PICH-deficient mouse models with the Eµ-Myc transgenic mouse model, which recapitulates B-cell lymphoma development. We have observed that PICH deficiency delays the onset of MYC-induced lymphomas in Pich heterozygous females. Moreover, using a Pich conditional knockout mouse model, we have found that Pich deletion in adult mice improves the survival of Eµ-Myc transgenic mice. Notably, we show that Pich deletion in healthy adult mice is well tolerated, supporting PICH as a suitable target for anticancer therapies. Finally, we have corroborated these findings in two human Burkitt lymphoma cell lines and we have found that the death of cancer cells was accompanied by chromosomal instability. Based on these findings, we propose PICH as a potential therapeutic target for Burkitt lymphoma and for other cancers where PICH is overexpressed.


Subject(s)
Burkitt Lymphoma , Adult , Female , Animals , Humans , Mice , Burkitt Lymphoma/genetics , Cell Line , Chromosomal Instability , Disease Models, Animal , Mice, Knockout , Mice, Transgenic , DNA
2.
Circulation ; 147(1): 47-65, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36325906

ABSTRACT

BACKGROUND: The complex genetics underlying human cardiac disease is evidenced by its heterogenous manifestation, multigenic basis, and sporadic occurrence. These features have hampered disease modeling and mechanistic understanding. Here, we show that 2 structural cardiac diseases, left ventricular noncompaction (LVNC) and bicuspid aortic valve, can be caused by a set of inherited heterozygous gene mutations affecting the NOTCH ligand regulator MIB1 (MINDBOMB1) and cosegregating genes. METHODS: We used CRISPR-Cas9 gene editing to generate mice harboring a nonsense or a missense MIB1 mutation that are both found in LVNC families. We also generated mice separately carrying these MIB1 mutations plus 5 additional cosegregating variants in the ASXL3, APCDD1, TMX3, CEP192, and BCL7A genes identified in these LVNC families by whole exome sequencing. Histological, developmental, and functional analyses of these mouse models were carried out by echocardiography and cardiac magnetic resonance imaging, together with gene expression profiling by RNA sequencing of both selected engineered mouse models and human induced pluripotent stem cell-derived cardiomyocytes. Potential biochemical interactions were assayed in vitro by coimmunoprecipitation and Western blot. RESULTS: Mice homozygous for the MIB1 nonsense mutation did not survive, and the mutation caused LVNC only in heteroallelic combination with a conditional allele inactivated in the myocardium. The heterozygous MIB1 missense allele leads to bicuspid aortic valve in a NOTCH-sensitized genetic background. These data suggest that development of LVNC is influenced by genetic modifiers present in affected families, whereas valve defects are highly sensitive to NOTCH haploinsufficiency. Whole exome sequencing of LVNC families revealed single-nucleotide gene variants of ASXL3, APCDD1, TMX3, CEP192, and BCL7A cosegregating with the MIB1 mutations and LVNC. In experiments with mice harboring the orthologous variants on the corresponding Mib1 backgrounds, triple heterozygous Mib1 Apcdd1 Asxl3 mice showed LVNC, whereas quadruple heterozygous Mib1 Cep192 Tmx3;Bcl7a mice developed bicuspid aortic valve and other valve-associated defects. Biochemical analysis suggested interactions between CEP192, BCL7A, and NOTCH. Gene expression profiling of mutant mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes revealed increased cardiomyocyte proliferation and defective morphological and metabolic maturation. CONCLUSIONS: These findings reveal a shared genetic substrate underlying LVNC and bicuspid aortic valve in which MIB1-NOTCH variants plays a crucial role in heterozygous combination with cosegregating genetic modifiers.


Subject(s)
Bicuspid Aortic Valve Disease , Cardiomyopathies , Heart Defects, Congenital , Induced Pluripotent Stem Cells , Humans , Animals , Mice , Heart Defects, Congenital/complications , Cardiomyopathies/etiology , Myocytes, Cardiac , Aortic Valve/diagnostic imaging , Transcription Factors , Chromosomal Proteins, Non-Histone
3.
Aging (Albany NY) ; 12(7): 5612-5624, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32253367

ABSTRACT

Replication Stress (RS) is a type of DNA damage generated at the replication fork, characterized by single-stranded DNA (ssDNA) accumulation, and which can be caused by a variety of factors. Previous studies have reported elevated RS levels in aged cells. In addition, mouse models with a deficient RS response show accelerated aging. However, the relevance of endogenous or physiological RS, compared to other sources of genomic instability, for the normal onset of aging is unknown. We have performed long term survival studies of transgenic mice with extra copies of the Chk1 and/or Rrm2 genes, which we previously showed extend the lifespan of a progeroid ATR-hypomorphic model suffering from high levels of RS. In contrast to their effect in the context of progeria, the lifespan of Chk1, Rrm2 and Chk1/Rrm2 transgenic mice was similar to WT littermates in physiological settings. Most mice studied died due to tumors -mainly lymphomas- irrespective of their genetic background. Interestingly, a higher but not statistically significant percentage of transgenic mice developed tumors compared to WT mice. Our results indicate that supraphysiological protection from RS does not extend lifespan, indicating that RS may not be a relevant source of genomic instability on the onset of normal aging.


Subject(s)
Checkpoint Kinase 1/genetics , DNA Damage , Longevity/genetics , Ribonucleoside Diphosphate Reductase/genetics , Animals , Checkpoint Kinase 1/metabolism , DNA Replication , Mice , Mice, Transgenic , Ribonucleoside Diphosphate Reductase/metabolism
4.
Cell Rep ; 24(12): 3274-3284, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30232008

ABSTRACT

PICH is a DNA translocase necessary for the resolution of ultrafine anaphase DNA bridges and to ensure the fidelity of chromosomal segregation. Here, we report the generation of an animal model deficient for PICH that allowed us to investigate its physiological relevance. Pich KO mice lose viability during embryonic development due to a global accumulation of DNA damage. However, despite the presence of chromosomal instability, extensive p53 activation, and increased apoptosis throughout the embryo, Pich KO embryos survive until day 12.5 of embryonic development. The absence of p53 failed to improve the viability of the Pich KO embryos, suggesting that the observed developmental defects are not solely due to p53-induced apoptosis. Moreover, Pich-deficient mouse embryonic fibroblasts exhibit chromosomal instability and are resistant to RASV12/E1A-induced transformation. Overall, our data indicate that PICH is essential to preserve chromosomal integrity in rapidly proliferating cells and is therefore critical during embryonic development and tumorigenesis.


Subject(s)
Chromosomal Instability , Embryonic Development/genetics , Animals , Apoptosis , Cells, Cultured , DNA Damage , DNA Helicases/metabolism , Mice , Tumor Suppressor Protein p53/metabolism
5.
Transgenic Res ; 26(3): 429-434, 2017 06.
Article in English | MEDLINE | ID: mdl-28105543

ABSTRACT

The generation of genetically engineered mouse models (GEMMs), including knock-out (KO) and knock-in (KI) models, often requires genomic screening of many mouse ES cell (mESC) clones by Southern blot. The use of large targeting constructs facilitates the recombination of exogenous DNA in a specific genomic locus, but limits the detection of its correct genomic integration by standard PCR methods. Genomic Long Range PCR (LR-PCR), using primers adjacent to the homology arms, has been used as an alternative to radioactive-based Southern blot screenings. However, LR-PCRs are often difficult and render many false positive and false negative results. Here, we propose an alternative screening method based on the detection of a genetic modification at the mRNA level, which we successfully optimized in two mouse models. This screening method consists of a reverse-transcription PCR (RT-PCR) using primers that match exons flanking the targeting construct. The detection of the expected modification in this PCR product confirms the integration at the correct genomic location and shows that the mutant mRNA is expressed. This is a simple and sensitive strategy to screen locus-specific recombination of targeting constructs which can also be useful to screen KO and KI mutant mice or cell lines including those generated by CRISPR/Cas9.


Subject(s)
Embryonic Stem Cells/physiology , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Blotting, Southern , Genes, BRCA1 , Mice, Transgenic
6.
Cardiovasc Res ; 112(2): 568-580, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27496872

ABSTRACT

AIM: To determine the role of NOTCH during the arterial injury response and the subsequent chronic arterial-wall inflammation underlying atherosclerosis. METHODS AND RESULTS: We have generated a mouse model of endothelial-specific (Cdh5-driven) depletion of the Notch effector recombination signal binding protein for immunoglobulin kappa J region (RBPJ) [(ApoE-/-); homozygous RBPJk conditional mice (RBPJflox/flox); Cadherin 5-CreERT, tamoxifen inducible driver mice (Cdh5-CreERT)]. Endothelial-specific deletion of RBPJ or systemic deletion of Notch1 in athero-susceptible ApoE-/- mice fed a high-cholesterol diet for 6 weeks resulted in reduced atherosclerosis in the aortic arch and sinus. Intravital microscopy revealed decreased leucocyte rolling on the endothelium of ApoE-/-; RBPJflox/flox; Cdh5-CreERT mice, correlating with a lowered content of leucocytes and macrophages in the vascular wall. Transcriptome analysis revealed down-regulation of proinflammatory and endothelial activation pathways in atherosclerotic tissue of RBPJ-mutant mice. During normal Notch activation, Jagged1 signalling up-regulation in endothelial cells promotes nuclear translocation of the Notch1 intracellular domain (N1ICD) and its physical interaction with nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). This N1ICD-NF-κB interaction is required for reciprocal transactivation of target genes, including vascular cell adhesion molecule-1. CONCLUSIONS: Notch signalling pathway inactivation decreases leucocyte rolling, thereby preventing endothelial dysfunction and vascular inflammation. Attenuation of Notch signalling might provide a treatment strategy for atherosclerosis.

7.
Circ Res ; 118(10): 1480-97, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27056911

ABSTRACT

RATIONALE: The Notch signaling pathway is crucial for primitive cardiac valve formation by epithelial-mesenchymal transition, and NOTCH1 mutations cause bicuspid aortic valve; however, the temporal requirement for the various Notch ligands and receptors during valve ontogeny is poorly understood. OBJECTIVE: The aim of this study is to determine the functional specificity of Notch in valve development. METHODS AND RESULTS: Using cardiac-specific conditional targeted mutant mice, we find that endothelial/endocardial deletion of Mib1-Dll4-Notch1 signaling, possibly favored by Manic-Fringe, is specifically required for cardiac epithelial-mesenchymal transition. Mice lacking endocardial Jag1, Notch1, or RBPJ displayed enlarged valve cusps, bicuspid aortic valve, and septal defects, indicating that endocardial Jag1 to Notch1 signaling is required for post-epithelial-mesenchymal transition valvulogenesis. Valve dysmorphology was associated with increased mesenchyme proliferation, indicating that Jag1-Notch1 signaling restricts mesenchyme cell proliferation non-cell autonomously. Gene profiling revealed upregulated Bmp signaling in Jag1-mutant valves, providing a molecular basis for the hyperproliferative phenotype. Significantly, the negative regulator of mesenchyme proliferation, Hbegf, was markedly reduced in Jag1-mutant valves. Hbegf expression in embryonic endocardial cells could be readily activated through a RBPJ-binding site, identifying Hbegf as an endocardial Notch target. Accordingly, addition of soluble heparin-binding EGF-like growth factor to Jag1-mutant outflow tract explant cultures rescued the hyperproliferative phenotype. CONCLUSIONS: During cardiac valve formation, Dll4-Notch1 signaling leads to epithelial-mesenchymal transition and cushion formation. Jag1-Notch1 signaling subsequently restrains Bmp-mediated valve mesenchyme proliferation by sustaining Hbegf-EGF receptor signaling. Our studies identify a mechanism of signaling cross talk during valve morphogenesis involved in the origin of congenital heart defects associated with reduced NOTCH function.


Subject(s)
Mitral Valve/metabolism , Morphogenesis , Receptor, Notch1/genetics , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Heparin-binding EGF-like Growth Factor/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitral Valve/abnormalities , Mitral Valve/embryology , Receptor, Notch1/metabolism , Up-Regulation
8.
Proc Natl Acad Sci U S A ; 111(35): 12925-9, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25139994

ABSTRACT

Conflicting results have been reported for the roles of cGMP and cGMP-dependent protein kinase I (cGKI) in various pathological conditions leading to cardiac hypertrophy and fibrosis. A cardioprotective effect of cGMP/cGKI has been reported in whole animals and isolated cardiomyocytes, but recent evidence from a mouse model expressing cGKIß only in smooth muscle (ßRM) but not in cardiomyocytes, endothelial cells, or fibroblasts has forced a reevaluation of the requirement for cGKI activity in the cardiomyocyte antihypertrophic effects of cGMP. In particular, ßRM mice developed the same hypertrophy as WT controls when subjected to thoracic aortic constriction or isoproterenol infusion. Here, we challenged ßRM and WT (Ctr) littermate control mice with angiotensin II (AII) infusion (7 d; 2 mg ⋅ kg(-1) ⋅ d(-1)) to induce hypertrophy. Both genotypes developed cardiac hypertrophy, which was more pronounced in Ctr animals. Cardiomyocyte size and interstitial fibrosis were increased equally in both genotypes. Addition of sildenafil, a phosphodiesterase 5 (PDE5) inhibitor, in the drinking water had a small effect in reducing myocyte hypertrophy in WT mice and no effect in ßRM mice. However, sildenafil substantially blocked the increase in collagen I, fibronectin 1, TGFß, and CTGF mRNA in Ctr but not in ßRM hearts. These data indicate that, for the initial phase of AII-induced cardiac hypertrophy, lack of cardiomyocyte cGKI activity does not worsen hypertrophic growth. However, expression of cGKI in one or more cell types other than smooth muscle is necessary to allow the antifibrotic effect of sildenafil.


Subject(s)
Angiotensin II/pharmacology , Cardiomegaly/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Hypertension/metabolism , Animals , Cardiomegaly/chemically induced , Cyclic GMP/metabolism , Fibrosis/chemically induced , Fibrosis/metabolism , Genetic Markers , Hypertension/chemically induced , Mice , Muscle, Smooth/metabolism , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Sildenafil Citrate , Sulfones/pharmacology , Vasoconstrictor Agents/pharmacology
9.
Cardiovasc Res ; 101(1): 97-107, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24130190

ABSTRACT

AIMS: Melusin is a muscle-specific chaperone protein whose expression is required for a compensatory hypertrophy response to pressure overload. Here, we evaluated the consequences of melusin overexpression in the setting of myocardial infarction (MI) using a comprehensive multicentre approach. METHODS AND RESULTS: Mice overexpressing melusin in the heart (TG) and wild-type controls (WT) were subjected to permanent LAD ligation and both the acute response (Day 3) and subsequent remodelling (2 weeks) were examined. Mortality in wild-type mice was significant between Days 3 and 7, primarily due to cardiac rupture, but melusin's overexpression strongly reduced mortality (43.2% in wild-type vs. 27.3% in melusin-TG, P = 0.005). At Day 3 after MI, a time point preceding the mortality peak, TG hearts had increased heat shock protein 70 expression, increased ERK1/2 signalling, reduced cardiomyocyte hyper-contractility and inflammatory cell infiltrates, and increased matricellular protein expression in the infarcted area. At 2 weeks after MI, melusin overexpression conferred a favourable adaptive remodelling characterized by reduced left ventricle dilatation and better preserved contractility in the presence of a comparable degree of hypertrophy. Adaptive remodelling in melusin TG mice was characterized by reduced apoptosis and fibrosis as well as increased cardiomyocyte contractility. CONCLUSIONS: Consistent with its function as a chaperone protein, melusin overexpression exerts a dual protective action following MI reducing an array of maladaptive processes. In the early phase after MI, reduced inflammation and myocyte remodelling protect against cardiac rupture. Chronically, reduced myocyte loss and matrix remodelling, with preserved myocyte contractility, confer adaptive LV remodelling.


Subject(s)
Cytoskeletal Proteins/metabolism , Muscle Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Ventricular Remodeling , Animals , Apoptosis , Collagen/metabolism , Excitation Contraction Coupling , Extracellular Matrix Proteins/metabolism , Female , Heart Rupture/etiology , Heart Rupture/metabolism , Heat-Shock Proteins/metabolism , Humans , Inflammation/metabolism , Male , Mice , Mice, Transgenic , Myocardial Contraction , Myocardial Infarction/complications
10.
Cell Mol Life Sci ; 70(21): 4047-54, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23532408

ABSTRACT

The ability of cardiomyocytes to detect mechanical and humoral stimuli is critical for adaptation of the myocardium in response to new conditions and for sustaining the increased workload during stress. While certain stimuli mediate a beneficial adaptation to stress conditions, others result in maladaptive remodelling, ultimately leading to heart failure. Specific signalling pathways activating either adaptive or maladaptive cardiac remodelling have been identified. Paradoxically, however, in a number of cases, the transduction pathways involved in such opposing responses engage the same signalling proteins. A notable example is the Raf-MEK1/2-ERK1/2 signalling pathway that can control both adaptive and maladaptive remodelling. ERK1/2 signalling requires a signalosome complex where a scaffold protein drives the assembly of these three kinases into a linear pathway to facilitate their sequential phosphorylation, ultimately targeting specific effector molecules. Interestingly, a number of different Raf-MEK1/2-ERK1/2 scaffold proteins have been identified, and their role in determining the adaptive or maladaptive cardiac remodelling is a promising field of investigation for the development of therapeutic strategies capable of selectively potentiating the adaptive response.


Subject(s)
Heart Failure/physiopathology , Heart/physiopathology , MAP Kinase Signaling System/physiology , Myocardium/pathology , Animals , Arrestins/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Humans , Mice , Myocytes, Cardiac/cytology , Phosphorylation , Protein Structure, Tertiary , Signal Transduction , beta-Arrestins , raf Kinases/metabolism
11.
Cell Cycle ; 10(21): 3678-83, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22037254

ABSTRACT

Chaperones and scaffold proteins are key elements involved in controlling the assembly of molecular complexes required for coordinated signal transduction. Here we describe morgana and melusin, two phylogenetically conserved chaperones that cooperate with Hsp90 and regulate signal transduction in important physiopathological processes. While morgana is ubiquitously expressed, melusin expression is restricted to striated muscles. Despite high sequence homology, the two chaperones have distinct functions. Morgana controls genomic stability by regulating the centrosome cycle via ROCKII kinase. Melusin, however, organizes ERK signal transduction in cardiomyocytes and regulates cardiac compensatory hypertrophy in response to different stress stimuli.


Subject(s)
Carrier Proteins/physiology , Cytoskeletal Proteins/physiology , Molecular Chaperones/physiology , Muscle Proteins/physiology , Signal Transduction , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Evolution, Molecular , Gene Duplication , Mice , Models, Molecular , Molecular Chaperones/chemistry , Muscle Proteins/chemistry , Muscle Proteins/genetics , Protein Structure, Tertiary
12.
J Cell Sci ; 124(Pt 20): 3515-24, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-22010199

ABSTRACT

Extracellular signal-regulated kinase 1/2 (ERK1/2) signalling is a key pathway in cardiomyocyte hypertrophy and survival in response to many different stress stimuli. We have previously characterized melusin as a muscle-specific chaperone protein capable of ERK1/2 signalling activation in the heart. Here, we show that in the heart, melusin forms a supramolecular complex with the proto-oncogene c-Raf, MEK1/2 (also known as MAPKK1/2) and ERK1/2 and that melusin-bound mitogen-activated protein kinases (MAPKs) are activated by pressure overload. Moreover, we demonstrate that both focal adhesion kinase (FAK) and IQ motif-containing GTPase activating protein 1 (IQGAP1), a scaffold protein for the ERK1/2 signalling cascade, are part of the melusin complex and are required for ERK1/2 activation in response to pressure overload. Finally, analysis of isolated neonatal cardiomyocytes indicates that both FAK and IQGAP1 regulate melusin-dependent cardiomyocyte hypertrophy and survival through ERK1/2 activation.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Cytoskeletal Proteins/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Molecular Chaperones/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , ras GTPase-Activating Proteins/metabolism , Allosteric Regulation , Animals , Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/physiopathology , Cell Survival/drug effects , Cells, Cultured , Cytoskeletal Proteins/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart/drug effects , Heart/physiology , Heart/physiopathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Mice, Transgenic , Molecular Chaperones/genetics , Multienzyme Complexes/metabolism , Muscle Proteins/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Stress, Physiological , ras GTPase-Activating Proteins/genetics
13.
Circulation ; 124(12): 1337-50, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21900081

ABSTRACT

BACKGROUND: Heart failure is one of the leading causes of mortality and is primarily the final stage of several overload cardiomyopathies, preceded by an early adaptive hypertrophic response and characterized by coordinated cardiomyocyte growth, angiogenesis, and inflammation. Therefore, growth factors and cytokines have to be critically regulated during cardiac response to transverse aortic constriction. Interestingly, the dual properties of placental growth factor as an angiogenic factor and cytokine make it a candidate to participate in cardiac remodeling in response to hemodynamic overload. METHODS AND RESULTS: After transverse aortic constriction, placental growth factor knockout mice displayed a dysregulation of cardiac remodeling, negatively affecting muscle growth. Molecular insights underscored that this effect was ascribable mainly to a failure in the establishment of adequate inflammatory response owing to an impaired activity of tumor necrosis factor-α-converting enzyme. Interestingly, after transverse aortic constriction, placental growth factor knockout mice had strongly increased levels of tissue inhibitor of metalloproteinases-3, the main natural TACE inhibitor, thus indicating an unbalance of the tissue inhibitor of metalloproteinases-3/tumor necrosis factor-α-converting enzyme axis. Strikingly, when we used an in vivo RNA interference approach to reduce tissue inhibitor of metalloproteinases-3 levels in placental growth factor knockout mice during transverse aortic constriction, we obtained a complete phenotype rescue of early dilated cardiomyopathy. CONCLUSIONS: Our results demonstrate that placental growth factor finely tunes a balanced regulation of the tissue inhibitor of metalloproteinases-3/tumor necrosis factor-α-converting enzyme axis and the consequent TNF-α activation in response to transverse aortic constriction, thus allowing the establishment of an inflammatory response necessary for adaptive cardiac remodeling.


Subject(s)
ADAM Proteins/metabolism , Hypertrophy, Left Ventricular/physiopathology , Myocarditis/physiopathology , Pregnancy Proteins/physiology , Tissue Inhibitor of Metalloproteinase-3/metabolism , Ventricular Remodeling/physiology , ADAM Proteins/physiology , ADAM17 Protein , Adaptation, Physiological/drug effects , Adaptation, Physiological/physiology , Animals , Aorta/physiopathology , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/physiopathology , Coronary Vessels/physiopathology , Disease Models, Animal , Hypertrophy, Left Ventricular/drug therapy , Male , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/drug therapy , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Placenta Growth Factor , Pregnancy Proteins/genetics , Pregnancy Proteins/pharmacology , Tissue Inhibitor of Metalloproteinase-3/physiology , Up-Regulation/drug effects , Up-Regulation/physiology , Ventricular Pressure/drug effects , Ventricular Pressure/physiology , Ventricular Remodeling/drug effects
14.
J Biol Chem ; 286(35): 30837-30846, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21757757

ABSTRACT

Maintenance of cardiac structure and Z-disc signaling are key factors responsible for protecting the heart in a setting of stress, but how these processes are regulated is not well defined. We recently demonstrated that PI3K(p110α) protects the heart against myocardial infarction. The aim of this study was to determine whether PI3K(p110α) directly regulates components of the Z-disc and cardiac structure. To address this question, a unique three-dimensional virtual muscle model was applied to gene expression data from transgenic mice with increased or decreased PI3K(p110α) activity under basal conditions (sham) and in a setting of myocardial infarction to display the location of structural proteins. Key findings from this analysis were then validated experimentally. The three-dimensional virtual muscle model visually highlighted reciprocally regulated transcripts associated with PI3K activation that encoded key components of the Z-disc and costamere, including melusin. Studies were performed to assess whether PI3K and melusin interact in the heart. Here, we identify a novel melusin-PI3K interaction that generates lipid kinase activity. The direct impact of PI3K(p110α) on myocyte structure was assessed by treating neonatal rat ventricular myocytes with PI3K(p110α) inhibitors and examining the myofiber morphology of hearts from PI3K transgenic mice. Results demonstrate that PI3K is critical for myofiber maturation and Z-disc alignment. In summary, PI3K regulates the expression of genes essential for cardiac structure and Z-disc signaling, interacts with melusin, and is critical for Z-disc alignment.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Gene Expression Regulation, Enzymologic , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Animals , Costameres/metabolism , Cytoskeletal Proteins/chemistry , Heart Failure/metabolism , Immunoprecipitation , Insulin Receptor Substrate Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal/methods , Muscle Cells/cytology , Muscle Proteins/chemistry , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism
15.
Cardiovasc Res ; 91(3): 456-64, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21493702

ABSTRACT

AIMS: The Raf-MEK1/2-ERK1/2 (ERK1/2-extracellular signal-regulated kinases 1/2) signalling cascade is crucial in triggering cardiac responses to different stress stimuli. Scaffold proteins are key elements in coordinating signalling molecules for their appropriate spatiotemporal activation. Here, we investigated the role of IQ motif-containing GTPase-activating protein 1 (IQGAP1), a scaffold for the ERK1/2 cascade, in heart function and remodelling in response to pressure overload. METHODS AND RESULTS: IQGAP1-null mice have unaltered basal heart function. When subjected to pressure overload, IQGAP1-null mice initially develop a compensatory hypertrophy indistinguishable from that of wild-type (WT) mice. However, upon a prolonged stimulus, the hypertrophic response develops towards a thinning of left ventricular walls, chamber dilation, and a decrease in contractility, in an accelerated fashion compared with WT mice. This unfavourable cardiac remodelling is characterized by blunted reactivation of the foetal gene programme, impaired cardiomyocyte hypertrophy, and increased cardiomyocyte apoptosis. Analysis of signalling pathways revealed two temporally distinct waves of both ERK1/2 and AKT phosphorylation peaking, respectively, at 10 min and 4 days after aortic banding in WT hearts. IQGAP1-null mice show strongly impaired phosphorylation of MEK1/2-ERK1/2 and AKT following 4 days of pressure overload, but normal activation of these kinases after 10 min. Pull-down experiments indicated that IQGAP1 is able to bind the three components of the ERK cascade, namely c-Raf, MEK1/2, and ERK1/2, as well as AKT in the heart. CONCLUSION: These data demonstrate, for the first time, a key role for the scaffold protein IQGAP1 in integrating hypertrophy and survival signals in the heart and regulating long-term left ventricle remodelling upon pressure overload.


Subject(s)
Blood Pressure , Hypertension/complications , Hypertrophy, Left Ventricular/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Ventricular Remodeling , ras GTPase-Activating Proteins/metabolism , Animals , Aorta/physiopathology , Aorta/surgery , Apoptosis , Cells, Cultured , Disease Models, Animal , Hypertension/enzymology , Hypertension/genetics , Hypertension/physiopathology , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Ligation , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Mice , Mice, 129 Strain , Mice, Knockout , Myocardium/pathology , Proto-Oncogene Proteins c-raf/metabolism , Time Factors , Ultrasonography , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
16.
Dev Cell ; 18(3): 486-95, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20230755

ABSTRACT

Centrosome abnormalities lead to genomic instability and are a common feature of many cancer cells. Here we show that mutations in morgana/chp-1 result in centrosome amplification and lethality in both Drosophila and mouse, and that the fly centrosome phenotype is fully rescued by the human ortholog of morgana. In mouse cells, morgana forms a complex with Hsp90 and ROCK I and II, and directly binds ROCK II. Morgana downregulation promotes the interaction between ROCK II and nucleophosmin (NPM), leading to an increased ROCK II kinase activity, which results in centrosome amplification. Morgana(+/-) primary cells and mice display an increased susceptibility to neoplastic transformation. In addition, tumor tissue array histochemical analysis revealed that morgana is underexpressed in a large fraction of breast and lung human cancers. Thus, morgana/chp-1 appears to prevent both centrosome amplification and tumorigenesis.


Subject(s)
Carrier Proteins/metabolism , Centrosome/metabolism , Centrosome/pathology , rho-Associated Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/genetics , Carrier Proteins/genetics , Cell Transformation, Neoplastic , Down-Regulation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryonic Development/genetics , Embryonic Development/physiology , Enzyme Inhibitors/metabolism , Female , Fungal Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Knockout , Mitosis/genetics , Mitosis/physiology , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Neoplasms, Experimental/etiology , Nuclear Proteins/metabolism , Nucleophosmin , Pregnancy
17.
FEBS Lett ; 582(13): 1788-94, 2008 Jun 11.
Article in English | MEDLINE | ID: mdl-18474241

ABSTRACT

Melusin is a mammalian muscle specific CHORD containing protein capable of activating signal transduction pathways leading to cardiomyocytes hypertrophy in response to mechanical stress. To define melusin function we searched for molecular partners possibly involved in melusin dependent signal transduction. Here we show that melusin and heat shock proteins are co-regulated. Moreover, melusin directly binds to Hsp90, a ubiquitous chaperone involved in regulating several signaling pathways. In addition, melusin interacts with Sgt1, an Hsp90 binding molecule. Melusin does not behave as an Hsp90 substrate but rather as a chaperone capable to protect citrate synthase from heat induced aggregation. These results describe melusin as a new component of the Hsp90 chaperone machinery.


Subject(s)
Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Muscle Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation , HSP90 Heat-Shock Proteins/genetics , Immunoprecipitation , Mice , Molecular Chaperones/genetics , Muscle Proteins/genetics , Protein Structure, Tertiary
18.
Circ Res ; 98(2): 218-25, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16357302

ABSTRACT

Diabetes mellitus is a main risk factor for vascular diseases. Vascular injury induced by diabetes mellitus is characterized by endothelial dysfunction attributable to an increased oxidative stress. So far, the molecular mechanisms involved in the vasculotoxic effects of diabetes are only partially known. We examined the effect of diabetes mellitus on oxidative stress and Rac-1 activation, a small G-protein involved in the activation of NADPH oxidase. Our results show that oxidative stress in vessels of different murine models of diabetes mellitus and in endothelial cells treated with high glucose is associated with an increased Rac-1/PAK binding and Rac-1 translocation from cytosol to plasma membrane, thus demonstrating an enhanced Rac-1 activity. More important, selective Rac-1 inhibition by an adenoviral vector carrying a dominant negative mutant of Rac-1 protected from oxidative stress and vascular dysfunction induced by diabetes mellitus. Our study demonstrates that Rac-1 plays a crucial role in diabetes-induced vascular injury, and it could be a target of novel therapeutic approaches to reduce vascular risk in diabetes mellitus.


Subject(s)
Diabetic Angiopathies/prevention & control , Neuropeptides/antagonists & inhibitors , rac GTP-Binding Proteins/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Animals , Cells, Cultured , Endothelium, Vascular/physiology , Glucose/pharmacology , Humans , Mice , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Neuropeptides/physiology , Oxidative Stress , Protein Kinase C/physiology , Protein Kinase C beta , rac GTP-Binding Proteins/physiology , rac1 GTP-Binding Protein/physiology
19.
Circ Res ; 96(10): 1087-94, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15860758

ABSTRACT

We have previously shown that genetic ablation of melusin, a muscle specific beta 1 integrin interacting protein, accelerates left ventricle (LV) dilation and heart failure in response to pressure overload. Here we show that melusin expression was increased during compensated cardiac hypertrophy in mice subjected to 1 week pressure overload, but returned to basal levels in LV that have undergone dilation after 12 weeks of pressure overload. To better understand the role of melusin in cardiac remodeling, we overexpressed melusin in heart of transgenic mice. Echocardiography analysis indicated that melusin over-expression induced a mild cardiac hypertrophy in basal conditions (30% increase in interventricular septum thickness) with no obvious structural and functional alterations. After prolonged pressure overload (12 weeks), melusin overexpressing hearts underwent further hypertrophy retaining concentric LV remodeling and full contractile function, whereas wild-type LV showed pronounced chamber dilation with an impaired contractility. Analysis of signaling pathways indicated that melusin overexpression induced increased basal phosphorylation of GSK3beta and ERK1/2. Moreover, AKT, GSK3beta and ERK1/2 were hyper-phosphorylated on pressure overload in melusin overexpressing compared with wild-type mice. In addition, after 12 weeks of pressure overload LV of melusin overexpressing mice showed a very low level of cardiomyocyte apoptosis and stromal tissue deposition, as well as increased capillary density compared with wild-type. These results demonstrate that melusin overexpression allows prolonged concentric compensatory hypertrophy and protects against the transition toward cardiac dilation and failure in response to long-standing pressure overload.


Subject(s)
Cardiomyopathy, Dilated/prevention & control , Cytoskeletal Proteins/physiology , Muscle Proteins/physiology , Myocardium/metabolism , Animals , Apoptosis , Blood Pressure , Cardiomyopathy, Dilated/etiology , Cytoskeletal Proteins/genetics , Fibrosis , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Hypertrophy, Left Ventricular/etiology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Muscle Proteins/genetics , Myocardium/pathology , Myocytes, Cardiac/pathology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...