Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
5.
Nat Commun ; 10(1): 329, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30659190

ABSTRACT

We previously demonstrated that beta II protein kinase C (ßIIPKC) activity is elevated in failing hearts and contributes to this pathology. Here we report that ßIIPKC accumulates on the mitochondrial outer membrane and phosphorylates mitofusin 1 (Mfn1) at serine 86. Mfn1 phosphorylation results in partial loss of its GTPase activity and in a buildup of fragmented and dysfunctional mitochondria in heart failure. ßIIPKC siRNA or a ßIIPKC inhibitor mitigates mitochondrial fragmentation and cell death. We confirm that Mfn1-ßIIPKC interaction alone is critical in inhibiting mitochondrial function and cardiac myocyte viability using SAMßA, a rationally-designed peptide that selectively antagonizes Mfn1-ßIIPKC association. SAMßA treatment protects cultured neonatal and adult cardiac myocytes, but not Mfn1 knockout cells, from stress-induced death. Importantly, SAMßA treatment re-establishes mitochondrial morphology and function and improves cardiac contractility in rats with heart failure, suggesting that SAMßA may be a potential treatment for patients with heart failure.


Subject(s)
Heart Failure/drug therapy , Membrane Proteins/antagonists & inhibitors , Mitochondrial Proteins/antagonists & inhibitors , Peptides/pharmacology , Protein Kinase C beta/antagonists & inhibitors , Animals , GTP Phosphohydrolases/metabolism , Gene Knockout Techniques , Heart Failure/metabolism , Male , Mitochondrial Membranes/metabolism , Myocardial Contraction , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Phosphorylation , RNA, Small Interfering , Rats, Wistar
6.
Cardiovasc Res ; 114(7): 1006-1015, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29579152

ABSTRACT

Aims: We previously demonstrated that acute ethanol administration protects the heart from ischaemia/reperfusion (I/R) injury thorough activation of aldehyde dehydrogenase 2 (ALDH2). Here, we characterized the role of acetaldehyde, an intermediate product from ethanol metabolism, and its metabolizing enzyme, ALDH2, in an ex vivo model of cardiac I/R injury. Methods and results: We used a combination of homozygous knock-in mice (ALDH2*2), carrying the human inactivating point mutation ALDH2 (E487K), and a direct activator of ALDH2, Alda-1, to investigate the cardiac effect of acetaldehyde. The ALDH2*2 mice have impaired acetaldehyde clearance, recapitulating the human phenotype. Yet, we found a similar infarct size in wild type (WT) and ALDH2*2 mice. Similar to ethanol-induced preconditioning, pre-treatment with 50 µM acetaldehyde increased ALDH2 activity and reduced cardiac injury in hearts of WT mice without affecting cardiac acetaldehyde levels. However, acetaldehyde pre-treatment of hearts of ALDH2*2 mice resulted in a three-fold increase in cardiac acetaldehyde levels and exacerbated I/R injury. Therefore, exogenous acetaldehyde appears to have a bimodal effect in I/R, depending on the ALDH2 genotype. Further supporting an ALDH2 role in cardiac preconditioning, pharmacological ALDH2 inhibition abolished ethanol-induced cardioprotection in hearts of WT mice, whereas a selective activator, Alda-1, protected ALDH2*2 against ethanol-induced cardiotoxicity. Finally, either genetic or pharmacological inhibition of ALDH2 mitigated ischaemic preconditioning. Conclusion: Taken together, our findings suggest that low levels of acetaldehyde are cardioprotective whereas high levels are damaging in an ex vivo model of I/R injury and that ALDH2 is a major, but not the only, regulator of cardiac acetaldehyde levels and protection from I/R.


Subject(s)
Acetaldehyde/pharmacology , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Aldehyde Dehydrogenase, Mitochondrial/genetics , Animals , Cardiotoxicity , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , Gene Knock-In Techniques , Genotype , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Phenotype , Point Mutation , Rats , Time Factors
7.
J Biomed Sci ; 24(1): 3, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-28056995

ABSTRACT

BACKGROUND: Mitochondrial aldehyde dehydrogenase 2 (ALDH2) is a key enzyme for the metabolism of many toxic aldehydes such as acetaldehyde, derived from alcohol drinking, and 4HNE, an oxidative stress-derived lipid peroxidation aldehyde. Post-translational enhancement of ALDH2 activity can be achieved by serine/threonine phosphorylation by epsilon protein kinase C (εPKC). Elevated ALDH2 is beneficial in reducing injury following myocardial infarction, stroke and other oxidative stress and aldehyde toxicity-related diseases. We have previously identified three εPKC phosphorylation sites, threonine 185 (T185), serine 279 (S279) and threonine 412 (T412), on ALDH2. Here we further characterized the role and contribution of each phosphorylation site to the enhancement of enzymatic activity by εPKC. METHODS: Each individual phosphorylation site was mutated to a negatively charged amino acid, glutamate, to mimic a phosphorylation, or to a non-phosphorylatable amino acid, alanine. ALDH2 enzyme activities and protection against 4HNE inactivation were measured in the presence or absence of εPKC phosphorylation in vitro. Coevolution of ALDH2 and its εPKC phosphorylation sites was delineated by multiple sequence alignments among a diverse range of species and within the ALDH multigene family. RESULTS: We identified S279 as a critical εPKC phosphorylation site in the activation of ALDH2. The critical catalytic site, cysteine 302 (C302) of ALDH2 is susceptible to adduct formation by reactive aldehyde, 4HNE, which readily renders the enzyme inactive. We show that phosphomimetic mutations of T185E, S279E and T412E confer protection of ALDH2 against 4HNE-induced inactivation, indicating that phosphorylation on these three sites by εPKC likely also protects the enzyme against reactive aldehydes. Finally, we demonstrate that the three ALDH2 phosphorylation sites co-evolved with εPKC over a wide range of species. Alignment of 18 human ALDH isozymes, indicates that T185 and S279 are unique ALDH2, εPKC specific phosphorylation sites, while T412 is found in other ALDH isozymes. We further identified three highly conserved serine/threonine residues (T384, T433 and S471) in all 18 ALDH isozymes that may play an important phosphorylation-mediated regulatory role in this important family of detoxifying enzymes. CONCLUSION: εPKC phosphorylation and its coevolution with ALDH2 play an important role in the regulation and protection of ALDH2 enzyme activity.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/chemistry , Evolution, Molecular , Protein Kinase C-epsilon/chemistry , Aldehyde Dehydrogenase, Mitochondrial/genetics , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Humans , Phosphorylation/physiology , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism
8.
J Exp Med ; 213(12): 2655-2669, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27821553

ABSTRACT

Huntington's disease (HD) is a rare genetic disease caused by expanded polyglutamine repeats in the huntingtin protein resulting in selective neuronal loss. Although genetic testing readily identifies those who will be affected, current pharmacological treatments do not prevent or slow down disease progression. A major challenge is the slow clinical progression and the inability to biopsy the affected tissue, the brain, making it difficult to design short and effective proof of concept clinical trials to assess treatment benefit. In this study, we focus on identifying peripheral biomarkers that correlate with the progression of the disease and treatment benefit. We recently developed an inhibitor of pathological mitochondrial fragmentation, P110, to inhibit neurotoxicity in HD. Changes in levels of mitochondrial DNA (mtDNA) and inflammation markers in plasma, a product of DNA oxidation in urine, mutant huntingtin aggregates, and 4-hydroxynonenal adducts in muscle and skin tissues were all noted in HD R6/2 mice relative to wild-type mice. Importantly, P110 treatment effectively reduced the levels of these biomarkers. Finally, abnormal levels of mtDNA were also found in plasma of HD patients relative to control subjects. Therefore, we identified several potential peripheral biomarkers as candidates to assess HD progression and the benefit of intervention for future clinical trials.


Subject(s)
Biomarkers/blood , Biomarkers/urine , Disease Progression , Huntington Disease/diagnosis , Huntington Disease/therapy , Aldehydes/metabolism , Animals , Behavior, Animal , Body Fluids/metabolism , Brain/metabolism , Brain/pathology , Case-Control Studies , DNA Damage , DNA, Mitochondrial/blood , Humans , Huntington Disease/blood , Huntington Disease/urine , Inflammation/complications , Inflammation/pathology , Mice , Mitochondria/metabolism , Muscle, Skeletal/pathology , Oxidation-Reduction , Phenotype , Protein Aggregates
9.
J Am Chem Soc ; 138(24): 7626-35, 2016 06 22.
Article in English | MEDLINE | ID: mdl-27218445

ABSTRACT

Protein kinases regulate numerous cellular processes, including cell growth, metabolism, and cell death. Because the primary sequence and the three-dimensional structure of many kinases are highly similar, the development of selective inhibitors for only one kinase is challenging. Furthermore, many protein kinases are pleiotropic, mediating diverse and sometimes even opposing functions by phosphorylating multiple protein substrates. Here, we set out to develop an inhibitor of a selective protein kinase phosphorylation of only one of its substrates. Focusing on the pleiotropic delta protein kinase C (δPKC), we used a rational approach to identify a distal docking site on δPKC for its substrate, pyruvate dehydrogenase kinase (PDK). We reasoned that an inhibitor of PDK's docking should selectively inhibit the phosphorylation of only PDK without affecting phosphorylation of the other δPKC substrates. Our approach identified a selective inhibitor of PDK docking to δPKC with an in vitro Kd of ∼50 nM and reducing cardiac injury IC50 of ∼5 nM. This inhibitor, which did not affect the phosphorylation of other δPKC substrates even at 1 µM, demonstrated that PDK phosphorylation alone is critical for δPKC-mediated injury by heart attack. The approach we describe is likely applicable for the identification of other substrate-specific kinase inhibitors.


Subject(s)
Myocardial Reperfusion Injury/drug therapy , Peptides/chemical synthesis , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Male , Mice, Inbred BALB C , Molecular Docking Simulation , Myocardial Reperfusion Injury/enzymology , Peptides/chemistry , Peptides/therapeutic use , Peptides/toxicity , Phosphorylation , Protein Binding , Protein Kinase C-delta/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/toxicity , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Rats, Sprague-Dawley , Rats, Wistar , Recombinant Proteins , Substrate Specificity
10.
EMBO Mol Med ; 7(10): 1307-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26268247

ABSTRACT

Mitochondrial dysfunction is implicated in multiple neurodegenerative diseases. In order to maintain a healthy population of functional mitochondria in cells, defective mitochondria must be properly eliminated by lysosomal machinery in a process referred to as mitophagy. Here, we uncover a new molecular mechanism underlying mitophagy driven by glyceraldehyde-3-phosphate dehydrogenase (GAPDH) under the pathological condition of Huntington's disease (HD) caused by expansion of polyglutamine repeats. Expression of expanded polyglutamine tracts catalytically inactivates GAPDH (iGAPDH), which triggers its selective association with damaged mitochondria in several cell culture models of HD. Through this mechanism, iGAPDH serves as a signaling molecule to induce direct engulfment of damaged mitochondria into lysosomes (micro-mitophagy). However, abnormal interaction of mitochondrial GAPDH with long polyglutamine tracts stalled GAPDH-mediated mitophagy, leading to accumulation of damaged mitochondria, and increased cell death. We further demonstrated that overexpression of inactive GAPDH rescues this blunted process and enhances mitochondrial function and cell survival, indicating a role for GAPDH-driven mitophagy in the pathology of HD.


Subject(s)
Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+) , Huntington Disease/metabolism , Huntington Disease/physiopathology , Mitophagy , Animals , Cell Survival , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/biosynthesis , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/genetics , Humans , Huntingtin Protein , Mice , Mice, Knockout , Mitochondria/metabolism , Nerve Tissue Proteins , Nuclear Proteins , PC12 Cells , Polyglutamic Acid/metabolism , Rats
11.
Circ Res ; 116(11): 1783-99, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25999419

ABSTRACT

Reactive oxygen species (ROS) have been implicated in a variety of age-related diseases, including multiple cardiovascular disorders. However, translation of ROS scavengers (antioxidants) into the clinic has not been successful. These antioxidants grossly reduce total levels of cellular ROS including ROS that participate in physiological signaling. In this review, we challenge the traditional antioxidant therapeutic approach that targets ROS directly with novel approaches that improve mitochondrial functions to more effectively treat cardiovascular diseases.


Subject(s)
Antioxidants/therapeutic use , Cardiovascular Diseases/drug therapy , Mitochondria, Heart/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Animals , Cardiovascular Diseases/metabolism , Humans , Mitochondria, Heart/metabolism , Models, Biological , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
12.
J Mol Med (Berl) ; 93(3): 279-87, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25652199

ABSTRACT

The processes that control the number and shape of the mitochondria (mitochondrial dynamics) and the removal of damaged mitochondria (mitophagy) have been the subject of intense research. Recent work indicates that these processes may contribute to the pathology associated with cardiac diseases. This review describes some of the key proteins that regulate these processes and their potential as therapeutic targets for cardiac diseases.


Subject(s)
Cardiotonic Agents/pharmacology , Heart Diseases/drug therapy , Mitochondrial Dynamics , Mitophagy , Animals , Cardiotonic Agents/therapeutic use , Heart Diseases/pathology , Humans , Mitochondria, Heart/physiology
13.
Sci Signal ; 7(350): ra105, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25372052

ABSTRACT

Linear consensus motifs are short contiguous sequences of residues within a protein that can form recognition modules for protein interaction or catalytic modification. Protein kinase specificity and the matching of kinases to substrates have been mostly defined by phosphorylation sites that occur in linear consensus motifs. However, phosphorylation can also occur within sequences that do not match known linear consensus motifs recognized by kinases and within flexible loops. We report the identification of Thr(253) in α-tubulin as a site that is phosphorylated by protein kinase C ßI (PKCßI). Thr(253) is not part of a linear PKC consensus motif. Instead, Thr(253) occurs within a region on the surface of α-tubulin that resembles a PKC phosphorylation site consensus motif formed by basic residues in different parts of the protein, which come together in the folded protein to form the recognition motif for PKCßI. Mutations of these basic residues decreased substrate phosphorylation, confirming the presence of this "structurally formed" consensus motif and its importance for the protein kinase-substrate interaction. Analysis of previously reported protein kinase A (PKA) and PKC substrates identified sites within structurally formed consensus motifs in many substrates of these two kinase families. Thus, the concept of consensus phosphorylation site motif needs to be expanded to include sites within these structurally formed consensus motifs.


Subject(s)
Phosphotransferases/chemistry , Amino Acid Motifs , Animals , Catalysis , Cattle , Cyclic AMP-Dependent Protein Kinases/chemistry , Green Fluorescent Proteins/chemistry , HEK293 Cells , HeLa Cells , Humans , Lysine/chemistry , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Protein Folding , Protein Kinase C/chemistry , Threonine/chemistry , Tubulin/chemistry
14.
Rare Dis ; 2: e28637, 2014.
Article in English | MEDLINE | ID: mdl-25054095

ABSTRACT

Huntington disease is a rare neurodegenerative disease resulting from insertion and/or expansion of a polyglutamine repeats close to the N-terminal of the huntingtin protein. Although unequivocal genetic tests have been available for about 20 years, current pharmacological treatments do not prevent or slow down disease progression. Recent basic research identified potential novel drug targets for the treatment of Huntington disease. However, there are clear challenges in translating these discoveries into treatment strategies for these patients. The following is a brief discussion of these challenges using our recent experience as an example.

15.
Cardiovasc Res ; 103(4): 498-508, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24817685

ABSTRACT

AIMS: We previously demonstrated that pharmacological activation of mitochondrial aldehyde dehydrogenase 2 (ALDH2) protects the heart against acute ischaemia/reperfusion injury. Here, we determined the benefits of chronic activation of ALDH2 on the progression of heart failure (HF) using a post-myocardial infarction model. METHODS AND RESULTS: We showed that a 6-week treatment of myocardial infarction-induced HF rats with a selective ALDH2 activator (Alda-1), starting 4 weeks after myocardial infarction at a time when ventricular remodelling and cardiac dysfunction were present, improved cardiomyocyte shortening, cardiac function, left ventricular compliance and diastolic function under basal conditions, and after isoproterenol stimulation. Importantly, sustained Alda-1 treatment showed no toxicity and promoted a cardiac anti-remodelling effect by suppressing myocardial hypertrophy and fibrosis. Moreover, accumulation of 4-hydroxynonenal (4-HNE)-protein adducts and protein carbonyls seen in HF was not observed in Alda-1-treated rats, suggesting that increasing the activity of ALDH2 contributes to the reduction of aldehydic load in failing hearts. ALDH2 activation was associated with improved mitochondrial function, including elevated mitochondrial respiratory control ratios and reduced H2O2 release. Importantly, selective ALDH2 activation decreased mitochondrial Ca(2+)-induced permeability transition and cytochrome c release in failing hearts. Further supporting a mitochondrial mechanism for ALDH2, Alda-1 treatment preserved mitochondrial function upon in vitro aldehydic load. CONCLUSIONS: Selective activation of mitochondrial ALDH2 is sufficient to improve the HF outcome by reducing the toxic effects of aldehydic overload on mitochondrial bioenergetics and reactive oxygen species generation, suggesting that ALDH2 activators, such as Alda-1, have a potential therapeutic value for treating HF patients.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Heart Failure/enzymology , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Ventricular Remodeling/physiology , Aldehyde Dehydrogenase, Mitochondrial , Animals , Heart Failure/physiopathology , Male , Myocardial Contraction/physiology , Myocytes, Cardiac/enzymology , Rats, Wistar , Ventricular Function/physiology
16.
J Clin Invest ; 123(12): 5371-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24231356

ABSTRACT

Huntington's disease (HD) is the result of expression of a mutated Huntingtin protein (mtHtt), and is associated with a variety of cellular dysfunctions including excessive mitochondrial fission. Here, we tested whether inhibition of excessive mitochondrial fission prevents mtHtt-induced pathology. We developed a selective inhibitor (P110-TAT) of the mitochondrial fission protein dynamin-related protein 1 (DRP1). We found that P110-TAT inhibited mtHtt-induced excessive mitochondrial fragmentation, improved mitochondrial function, and increased cell viability in HD cell culture models. P110-TAT treatment of fibroblasts from patients with HD and patients with HD with iPS cell-derived neurons reduced mitochondrial fragmentation and corrected mitochondrial dysfunction. P110-TAT treatment also reduced the extent of neurite shortening and cell death in iPS cell-derived neurons in patients with HD. Moreover, treatment of HD transgenic mice with P110-TAT reduced mitochondrial dysfunction, motor deficits, neuropathology, and mortality. We found that p53, a stress gene involved in HD pathogenesis, binds to DRP1 and mediates DRP1-induced mitochondrial and neuronal damage. Furthermore, P110-TAT treatment suppressed mtHtt-induced association of p53 with mitochondria in multiple HD models. These data indicate that inhibition of DRP1-dependent excessive mitochondrial fission with a P110-TAT-like inhibitor may prevent or slow the progression of HD.


Subject(s)
Dynamins/antagonists & inhibitors , GTP Phosphohydrolases/antagonists & inhibitors , Huntington Disease/drug therapy , Microtubule-Associated Proteins/antagonists & inhibitors , Mitochondria/drug effects , Mitochondrial Proteins/antagonists & inhibitors , Adult , Animals , Cells, Cultured , Child , Corpus Striatum/pathology , Disease Models, Animal , Fibroblasts , Gene Knock-In Techniques , HEK293 Cells , Humans , Huntington Disease/pathology , Induced Pluripotent Stem Cells/pathology , Male , Mice , Mice, Transgenic , Mitochondria/physiology , Molecular Targeted Therapy , Nerve Degeneration/prevention & control , Neurites/ultrastructure , Neurogenesis , Protein Binding , Recombinant Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
17.
J Am Heart Assoc ; 2(5): e000461, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24103571

ABSTRACT

BACKGROUND: Ischemia and reperfusion (IR) injury remains a major cause of morbidity and mortality and multiple molecular and cellular pathways have been implicated in this injury. We determined whether acute inhibition of excessive mitochondrial fission at the onset of reperfusion improves mitochondrial dysfunction and cardiac contractility postmyocardial infarction in rats. METHODS AND RESULTS: We used a selective inhibitor of the fission machinery, P110, which we have recently designed. P110 treatment inhibited the interaction of fission proteins Fis1/Drp1, decreased mitochondrial fission, and improved bioenergetics in three different rat models of IR, including primary cardiomyocytes, ex vivo heart model, and an in vivo myocardial infarction model. Drp1 transiently bound to the mitochondria following IR injury and P110 treatment blocked this Drp1 mitochondrial association. Compared with control treatment, P110 (1 µmol/L) decreased infarct size by 28 ± 2% and increased adenosine triphosphate levels by 70+1% after IR relative to control IR in the ex vivo model. Intraperitoneal injection of P110 (0.5 mg/kg) at the onset of reperfusion in an in vivo model resulted in improved mitochondrial oxygen consumption by 68% when measured 3 weeks after ischemic injury, improved cardiac fractional shortening by 35%, reduced mitochondrial H2O2 uncoupling state by 70%, and improved overall mitochondrial functions. CONCLUSIONS: Together, we show that excessive mitochondrial fission at reperfusion contributes to long-term cardiac dysfunction in rats and that acute inhibition of excessive mitochondrial fission at the onset of reperfusion is sufficient to result in long-term benefits as evidenced by inhibiting cardiac dysfunction 3 weeks after acute myocardial infarction.


Subject(s)
GTP Phosphohydrolases/pharmacology , Mitochondria, Heart/drug effects , Mitochondria, Heart/physiology , Mitochondrial Dynamics/drug effects , Myocardial Infarction/physiopathology , Peptide Fragments/pharmacology , Animals , Cells, Cultured , Myocardial Infarction/drug therapy , Myocytes, Cardiac/drug effects , Rats , Time Factors
18.
J Mol Cell Cardiol ; 51(6): 980-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21920368

ABSTRACT

We previously found that in the hearts of hypertensive Dahl salt-sensitive rats, ßIIPKC levels increase during the transition from compensated cardiac hypertrophy to cardiac dysfunction. Here we showed that a six-week treatment of these hypertensive rats with a ßIIPKC-specific inhibitor, ßIIV5-3, prolonged their survival by at least 6weeks, suppressed myocardial fibrosis and inflammation, and delayed the transition from compensated hypertrophy to cardiac dysfunction. In addition, changes in the levels of the Ca(2+)-handling proteins, SERCA2 and the Na(+)/Ca(2+) exchanger, as well as troponin I phosphorylation, seen in the control-treated hypertensive rats were not observed in the ßΙΙPKC-treated rats, suggesting that ßΙΙPKC contributes to the regulation of calcium levels in the myocardium. In contrast, treatment with the selective inhibitor of ßIPKC, an alternative spliced form of ßIIPKC, had no beneficial effects in these rats. We also found that ßIIV5-3, but not ßIV5-3, improved calcium handling in isolated rat cardiomyocytes and enhanced contractility in isolated rat hearts. In conclusion, our data using an in vivo model of cardiac dysfunction (late-phase hypertrophy), suggest that ßIIPKC contributes to the pathology associated with heart failure and thus an inhibitor of ßIIPKC may be a potential treatment for this disease.


Subject(s)
Cardiomegaly/enzymology , Cardiomegaly/prevention & control , Peptides/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Amino Acid Sequence , Animals , Calcium/metabolism , Cardiomegaly/mortality , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Disease Models, Animal , Fibrosis , Heart Failure/metabolism , Heart Failure/mortality , Heart Failure/prevention & control , Male , Myocardial Contraction/drug effects , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Protein Kinase C beta , Rats , Rats, Inbred Dahl , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism
19.
Mol Biol Cell ; 22(2): 256-65, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21119009

ABSTRACT

Neuronal cell death in a number of neurological disorders is associated with aberrant mitochondrial dynamics and mitochondrial degeneration. However, the triggers for this mitochondrial dysregulation are not known. Here we show excessive mitochondrial fission and mitochondrial structural disarray in brains of hypertensive rats with hypertension-induced brain injury (encephalopathy). We found that activation of protein kinase Cδ (PKCδ) induced aberrant mitochondrial fragmentation and impaired mitochondrial function in cultured SH-SY5Y neuronal cells and in this rat model of hypertension-induced encephalopathy. Immunoprecipitation studies indicate that PKCδ binds Drp1, a major mitochondrial fission protein, and phosphorylates Drp1 at Ser 579, thus increasing mitochondrial fragmentation. Further, we found that Drp1 Ser 579 phosphorylation by PKCδ is associated with Drp1 translocation to the mitochondria under oxidative stress. Importantly, inhibition of PKCδ, using a selective PKCδ peptide inhibitor (δV1-1), reduced mitochondrial fission and fragmentation and conferred neuronal protection in vivo and in culture. Our study suggests that PKCδ activation dysregulates the mitochondrial fission machinery and induces aberrant mitochondrial fission, thus contributing to neurological pathology.


Subject(s)
Dynamins/metabolism , Mitochondria/metabolism , Oxidative Stress , Protein Kinase C-delta/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Brain/ultrastructure , Cell Death , Humans , Hypertension/metabolism , Hypertension/pathology , Male , Mitochondria/ultrastructure , Multiprotein Complexes/metabolism , Neurons/pathology , Oligopeptides/pharmacology , Phosphorylation , Protein Binding , Protein Kinase C-delta/antagonists & inhibitors , Protein Structure, Tertiary , Protein Transport , Rats , Rats, Inbred Dahl , Sequence Alignment , Tumor Cells, Cultured
20.
Cardiovasc Res ; 88(1): 83-92, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20558438

ABSTRACT

AIMS: Protein kinase C epsilon (PKCepsilon) is critical for cardiac protection from ischaemia and reperfusion (IR) injury. PKCepsilon substrates that mediate cytoprotection reside in the mitochondria. However, the mechanism enabling mitochondrial translocation and import of PKCepsilon to enable phosphorylation of these substrates is not known. Heat shock protein 90 (HSP90) is a cytoprotective protein chaperone that participates in mitochondrial import of a number of proteins. Here, we investigated the role of HSP90 in mitochondrial import of PKCepsilon. METHODS AND RESULTS: Using an ex vivo perfused rat heart model of IR, we found that PKCepsilon translocates from the cytosol to the mitochondrial fraction following IR. Immunogold electron microscopy and mitochondrial fractionation demonstrated that following IR, mitochondrial PKCepsilon is localized within the mitochondria, on the inner mitochondrial membrane. Pharmacological inhibition of HSP90 prevented IR-induced interaction between PKCepsilon and the translocase of the outer membrane (Tom20), reduced mitochondrial import of PKCepsilon, and increased necrotic cell death by approximately 70%. Using a rational approach, we designed a 7-amino acid peptide activator of PKCepsilon, derived from an HSP90 homologous sequence located in the C2 domain of PKCepsilon (termed psiepsilonHSP90). Treatment with this peptide (conjugated to the cell permeating TAT protein-derived peptide, TAT(47-57)) increased PKCepsilon-HSP90 protein-protein interaction, enhanced mitochondrial translocation of PKCepsilon, increased phosphorylation and activity of an intra-mitochondrial PKCepsilon substrate, aldehyde dehydrogenase 2, and reduced cardiac injury in ex vivo and in vivo models of myocardial infarction. CONCLUSION: Our results suggest that HSP90-mediated mitochondrial import of PKCepsilon plays an important role in the protection of the myocardium from IR injury.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Mitochondria, Heart/enzymology , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Protein Kinase C-epsilon/metabolism , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Amino Acid Sequence , Animals , Cytoprotection , Disease Models, Animal , Drug Design , Enzyme Activation , Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Humans , Male , Membrane Transport Proteins , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Mitochondrial Membranes/enzymology , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Myocardial Contraction , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Necrosis , Oligopeptides/chemistry , Oligopeptides/pharmacology , Phosphorylation , Protein Binding , Protein Transport , Rats , Rats, Wistar , Receptors, Cell Surface , Receptors, Cytoplasmic and Nuclear/metabolism , Recovery of Function , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...