Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
JCI Insight ; 52019 06 13.
Article in English | MEDLINE | ID: mdl-31194698

ABSTRACT

Arrhythmogenic cardiomyopathy (ACM) is an inherited disorder with variable genetic etiologies. Here we focused on understanding the precise molecular pathology of a single clinical variant in DSP, the gene encoding desmoplakin. We initially identified a novel missense desmoplakin variant (p.R451G) in a patient diagnosed with biventricular ACM. An extensive single-family ACM cohort was assembled, revealing a pattern of coinheritance for R451G desmoplakin and the ACM phenotype. An in vitro model system using patient-derived induced pluripotent stem cell lines showed depressed levels of desmoplakin in the absence of abnormal electrical propagation. Molecular dynamics simulations of desmoplakin R451G revealed no overt structural changes, but a significant loss of intramolecular interactions surrounding a putative calpain target site was observed. Protein degradation assays of recombinant desmoplakin R451G confirmed increased calpain vulnerability. In silico screening identified a subset of 3 additional ACM-linked desmoplakin missense mutations with apparent enhanced calpain susceptibility, predictions that were confirmed experimentally. Like R451G, these mutations are found in families with biventricular ACM. We conclude that augmented calpain-mediated degradation of desmoplakin represents a shared pathological mechanism for select ACM-linked missense variants. This approach for identifying variants with shared molecular pathologies may represent a powerful new strategy for understanding and treating inherited cardiomyopathies.


Subject(s)
Arrhythmias, Cardiac/genetics , Calpain/metabolism , Cardiomyopathies/genetics , Desmoplakins/metabolism , Genetic Predisposition to Disease/genetics , Mutation , Adult , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Calpain/pharmacology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Desmoplakins/antagonists & inhibitors , Desmoplakins/chemistry , Female , Glycine , Heart , Heart Failure , Humans , Male , Middle Aged , Models, Molecular , Mutagenesis, Site-Directed , Mutation, Missense , Pedigree , Phenotype , Recombinant Proteins , Stem Cells
2.
Theranostics ; 6(11): 1947-62, 2016.
Article in English | MEDLINE | ID: mdl-27570562

ABSTRACT

Quiescent leukemia stem cells (LSCs) that are insensitive to BCR-ABL tyrosine kinase inhibitors confer resistance to imatinib in chronic myelogenous leukemia (CML). Identifying proteins to regulate survival and stemness of LSCs is urgently needed. Although histone deacetylase inhibitors (HDACis) can eliminate quiescent LSCs in CML, little is known about the underlying mechanism that HDACis kill LSCs. By fishing with a biotin-labeled probe, we identified that HDACi JSL-1 bound to the protein γ-catenin. γ-Catenin expression was higher in LSCs from CML patients than normal hematopoietic stem cells. Silencing γ-catenin in human CML CD34(+) bone-marrow (BM) cells sufficiently eliminated LSCs, which suggests that γ-catenin is required for survival of CML LSCs. Pharmacological inhibition of γ-catenin thwarted survival and self-renewal of human CML CD34(+) cells in vitro, and of murine LSCs in BCR-ABL-driven CML mice. γ-Catenin inhibition reduced long-term engraftment of human CML CD34(+) cells in NOD.Cg-Prkdc (scid) II2rg (tm1Sug)/JicCrl (NOG) mice. Silencing γ-catenin by shRNA in human primary CD34(+) cells did not alter ß-catenin, implying a ß-catenin-independent role of γ-catenin in survival and self-renewal of CML LSCs. Taken together, our findings validate that γ-catenin may be a novel therapeutic target of LSCs, and suppression of γ-catenin by HDACi may explain elimination of CML LSCs.


Subject(s)
Antineoplastic Agents/administration & dosage , Desmoplakins/antagonists & inhibitors , Histone Deacetylase Inhibitors/administration & dosage , Imatinib Mesylate/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/drug effects , Animals , Cell Survival/drug effects , Disease Models, Animal , Heterografts , Humans , Mice , Treatment Outcome , Tumor Cells, Cultured , gamma Catenin
3.
Mol Med Rep ; 8(3): 780-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23877755

ABSTRACT

Desmosomes and gap junctions are situated in the intercalated disks of cardiac muscle and maintain the integrity of mechanical coupling and electrical impulse conduction between cells. The desmosomal plakin protein, desmoplakin (DSP), also plays a crucial role in the stability of these interconnected components as well as gap junction connexin proteins. In addition to cell­to­cell junctions, other molecules, including voltage­gated sodium channels (Nav1.5) are present in the intercalated disk and support the contraction of cardiac muscle. Mutations in genes encoding desmosome proteins may result in fatal arrhythmias, including arrhythmogenic right ventricular cardiomyopathy (ARVC). Therefore, the aim of the present study was to determine whether the presence of DSP is necessary for the normal function and localization of gap junction protein connexin43 (Cx43) and Nav1.5. To examine this hypothesis, RNA interference was utilized to knock down the expression of DSP in HL­1 cells and the content, distribution and function of Cx43 and Nav1.5 was assessed. Western blotting and flow cytometry experiments revealed that Cx43 and Nav1.5 expression decreased following DSP silencing. In addition, immunofluorescence studies demonstrated that a loss of DSP expression led to an abnormal distribution of Cx43 and Nav1.5, while scrape­loading dye/transfer revealed a decrease in dye transfer in DSP siRNA­treated cells. The sodium current was also recorded by the whole­cell patch clamp technique. The results indicated that DSP suppression decreased sodium current and slowed conduction velocity in cultured cells. The present study indicates that impaired mechanical coupling largely affects electrical synchrony, further uncovering the pathogenesis of ARVC.


Subject(s)
Connexin 43/metabolism , Desmoplakins/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sodium/metabolism , Animals , Cell Line , Desmoplakins/antagonists & inhibitors , Desmoplakins/metabolism , Gap Junctions/physiology , Membrane Potentials , Mice , Microscopy, Confocal , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , RNA Interference , RNA, Small Interfering/metabolism
4.
Exp Cell Res ; 313(11): 2336-44, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17475244

ABSTRACT

The intercellular adhesive junction desmosomes are essential for the maintenance of tissue structure and integrity in skin. Desmoplakin (Dp) is a major obligate plaque protein which plays a fundamental role in anchoring intermediate filaments to desmosomal cadherins. Evidence from hereditary human disease caused by mutations in the gene encoding Dp, e.g. Dp haploinsufficiency, suggests that alterations in Dp expression result not only in the disruption of tissue structure and integrity but also could evoke changes in keratinocyte proliferation. We have used transient RNA interference (RNAi) to downregulate Dp specifically in HaCaT keratinocytes. We showed that this Dp downregulation also caused reduced expression of several other desmosomal proteins. Increased cell proliferation and enhanced G(1)-to-S-phase entry in the cell cycle, as monitored by colonial cellular density and BrdU incorporation, were seen in Dp RNAi-treated cells. These proliferative changes were associated with elevated phospho-ERK1/2 and phospho-Akt levels. Furthermore, this increase in phospho-ERK/1/2 and phospho-Akt levels was sustained in Dp RNAi-treated cells at confluence whereas in control cells there was a significant reduction in phosphorylation of ERK1/2. This study indicates that Dp may participate in the regulation of keratinocyte cell proliferation by, in part at least, regulating cell cycle progression.


Subject(s)
Cell Cycle/genetics , Desmoplakins/physiology , Keratinocytes/physiology , Bromodeoxyuridine/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Desmoplakins/antagonists & inhibitors , Desmoplakins/genetics , Desmosomal Cadherins/metabolism , Desmosomes/drug effects , Desmosomes/genetics , Desmosomes/metabolism , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Keratinocytes/metabolism , Phosphorylation , Protein Precursors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...