Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Ther Innov Regul Sci ; 58(4): 655-662, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38530629

ABSTRACT

BACKGROUND: Appropriate exploratory efficacy data from Phase I trials are vital for subsequent phases. Owing to the uniqueness of brain tumors (BTs), use of different strategies to evaluate efficacy is warranted. We studied exploratory efficacy evaluation in Phase I trials involving BTs. METHODS: Using Clarivate's Cortellis™, 42 Phase I trials of BT interventions conducted from 2020 to 2022 were analyzed for efficacy endpoints, which were set as primary endpoints (PEs) or secondary endpoints (SEs). Additionally, these metrics were compared in two subgroups: trials including only BTs (Group-A) and those including BTs among mixed solid tumors (Group-B). RESULTS: Selected studies included a median of 1.5 PEs (range, 1-6) and 5 SEs (range, 0-19). Efficacy endpoints were included as PEs and SEs in 2 (5%) and 31 (78%) trials, respectively. Among the latter 31 trials that included 94 efficacy endpoints, 24, 22, 20, 9, and 8 reflected overall response rate (ORR), progression-free survival (PFS), overall survival (OS), duration of response (DOR), and disease control rate (DCR), respectively. ORR for BT was determined using various methods; however, the Response Evaluation Criteria in Solid Tumors (RECIST) was used less frequently in Group-A than in Group-B (p = 0.0039). CONCLUSIONS: Recent Phase I trials included efficacy endpoints as SEs, with ORR, PFS, or OS included in ~ 50% trials and DOR or DCR in ~ 25%. No established criteria exist for imaging evaluation of BTs. Phase I trials involving mixed solid tumor cohorts revealed challenges in designing methods to assess the exploratory efficacy of BTs.


Subject(s)
Brain Neoplasms , Clinical Trials, Phase I as Topic , Endpoint Determination , Humans , Brain Neoplasms/drug therapy , Progression-Free Survival , Treatment Outcome
2.
Ther Innov Regul Sci ; 57(3): 603-610, 2023 05.
Article in English | MEDLINE | ID: mdl-36602756

ABSTRACT

BACKGROUND: Response Evaluation Criteria in Solid Tumors (RECIST)-based response rates are commonly used as efficacy endpoints in phase II clinical trials for solid tumors. However, no consensus has been reached concerning adequate efficacy endpoints for phase II clinical trials targeting meningioma. Irregularity of lesions after resection, and varying degrees of dysplasia and histologic subtypes make establishing an appropriate efficacy evaluation difficult. METHODS: We analyzed primary efficacy endpoints (PEEs) and background factors from 48 trials retrieved from ClinicalTrials.gov ( https://clinicaltrials.gov/ ) using the search criteria "meningioma," "interventional," "phase II," and "study start 4/1/2001 to 3/31/2021." Primary purpose of the study was efficacy endpoint setting in overall population and three subgroups. RESULTS: Among 45 PEEs set in the 39 trials included; 33 trials with single PEE, and six trials with double PEEs, 17/45 (38%) trials adopted progression-free survival (PFS) rate, 15/45 (33%) trials response rate (seven Macdonald criteria or modified, three RECIST, three volumetric estimation, one RANO criteria, one unknown), 10/45 (22%) PFS, 1/45 (2%) OS, and 2/45 (4%) other endpoints. Although 26 PEEs were time-to-event endpoints, 19 of the 26 PEEs were single-arm studies. CONCLUSIONS: Time-to-event efficacy endpoints were often compared to historical data, and two-dimensional evaluation is more suitable than one-dimensional one. Accumulation of prognostic data is essential to standardize time-to-event efficacy endpoints. Considering the difficulty of setting design for phase II clinical studies targeting meningioma, evaluation might be done with multiple efficacy endpoints.


Subject(s)
Meningeal Neoplasms , Meningioma , Humans , Meningioma/drug therapy , Progression-Free Survival , Meningeal Neoplasms/drug therapy
3.
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
4.
Prostate ; 71(9): 946-54, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21541971

ABSTRACT

BACKGROUND: PKCδ is generally known as a pro-apoptotic and anti-proliferative enzyme in human prostate cancer cells. METHODS: Here, we investigated the role of PKCδ on the growth of PC-3 human prostate cancer cells in vivo and in vitro. RESULTS: We found that sustained treatment with a specific PKCδ activator (ψδ receptor for active C kinase, ψδRACK) increased growth of PC-3 xenografts. There was increased levels of HIF-1α, vascular endothelial growth factor and CD31-positive cells in PC-3 xenografts, representative of increased tumor angiogenesis. Mechanistically, PKCδ activation increased the levels of reactive oxygen species (ROS) by binding to and phosphorylating NADPH oxidase, which induced its activity. Also, PKCδ-induced activation of NADPH oxidase increased the level of HIF-1α. CONCLUSIONS: Our results using tumors from the PC-3 xenograft model suggest that PKCδ activation increases angiogenic activity in androgen-independent PC-3 prostate cancer cells by increasing NADPH oxidase activity and HIF-1α levels and thus may partly be responsible for increased angiogenesis in advanced prostate cancer.


Subject(s)
NADPH Oxidases/metabolism , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/enzymology , Protein Kinase C-delta/metabolism , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Enzyme Activation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Nude , Neovascularization, Pathologic/enzymology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Prostatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/metabolism
5.
Circulation ; 122(11 Suppl): S170-8, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20837910

ABSTRACT

BACKGROUND: ε protein kinase C (εPKC) is involved in vascular smooth muscle cell (VSMC) activation, but little is known about its function in vascular pathology. We aimed at assessing the role of εPKC in the development of restenosis. METHODS AND RESULTS: Rat models of aortic balloon injury with or without subsequent stenting were used. Rats were treated with the selective ψεPKC activator ε receptor for activated protein kinase C (ψεRACK), the selective εPKC inhibitor εV1-2, or saline. Both down-stream cascades of the platelet-derived growth factor receptor via extracellular signal-regulated kinase and Akt, respectively, were evaluated in vivo and in VSMC cultures. Intimal hyperplasia with luminal obliteration developed in saline-treated balloon-injured rat aortas (20.3±8.0%), and ψεRACK significantly promoted neointima development (32.4±4.9%, P=0.033), whereas εV1-2 significantly inhibited luminal narrowing (9.2±4.3%, P=0.039). εPKC inhibition led to significantly reduced VSMC extracellular signal-regulated kinase phosphorylation in vivo, whereas Akt phosphorylation was not markedly affected. Neointimal proliferation in vivo and platelet-derived growth factor-induced VSMC proliferation/migration in vitro were significantly inhibited by εV1-2. The inhibition of the platelet-derived growth factor pathway was mediated by inhibiting down-stream extracellular signal-regulated kinase and Akt phosphorylation. In vitro, εV1-2 showed inhibitory properties on endothelial cell proliferation, but that did not prevent reendothelialization in vivo. εV1-2 showed proapoptotic effects on VSMC in vitro. After stent implantation, luminal restenosis (quantified by optical coherence tomography imaging) was significantly reduced with εV1-2 (8.0±2.0%) compared with saline (20.2±9.8%, P=0.028). CONCLUSIONS: εPKC seems to be centrally involved in the development of neointimal hyperplasia. We suggest that εPKC inhibition may be mediated via inhibition of extracellular signal-regulated kinase and Akt activation. εPKC modulation may become a new therapeutic target against vascular restenosis.


Subject(s)
Aorta , Endothelial Cells/enzymology , Graft Occlusion, Vascular , Myocytes, Smooth Muscle/enzymology , Protein Kinase C-epsilon , Protein Kinase Inhibitors/pharmacology , Animals , Aorta/enzymology , Aorta/injuries , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation , Cells, Cultured , Coculture Techniques , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Graft Occlusion, Vascular/enzymology , Humans , Male , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
6.
Am J Physiol Heart Circ Physiol ; 295(1): H220-6, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18487446

ABSTRACT

Hypertension induced by high-salt diet in Dahl salt-sensitive rats leads to compensatory cardiac hypertrophy by approximately 11 wk, cardiac dysfunction at approximately 17 wk, and death from cardiac dysfunction at approximately 21 wk. It is unclear what molecular hallmarks distinguish the compensatory hypertrophy from the decompensated cardiac dysfunction phase. Here we compared the gene expression in rat cardiac tissue from the compensatory hypertrophic phase (11 wk, n = 6) with the cardiac dysfunction phase (17 wk, n = 6) and with age-matched normotensive controls. Messenger RNA levels of 93 genes, selected based on predicted association with cardiac dysfunction, were measured by quantitative real-time PCR. In the hypertrophic phase, the expression of three genes, atrial natriuretic peptide (ANP; P = 0.0089), brain natriuretic peptide (P = 0.0012), and endothelin-1 precursor (P = 0.028), significantly increased, whereas there was decreased expression of 24 other genes including SOD2 (P = 0.0148), sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a (P = 0.0002), and ryanodine receptor 2 (P = 0.0319). In the subsequent heart cardiac dysfunction phase, the expression of an additional 20 genes including inducible nitric oxide synthase (NOS; P = 0.0135), angiotensin I-converting enzyme (P = 0.0082), and IL-1beta (P < 0.0001) increased, whereas the expression of seven genes decreased compared with those of age-matched controls. Furthermore, the expression of 22 genes, including prepro-endothelin-1, ANP, angiotensin I-converting enzyme, beta(1)-adrenergic receptor, SOD2, and endothelial NOS, significantly changed in the cardiac dysfunction phase compared with the compensatory hypertrophic phase. Finally, principal component analysis successfully segregated animals with decompensatory cardiac dysfunction from controls, as well as from animals at the compensated hypertrophy phase, suggesting that we have identified molecular markers for each stage of the disease.


Subject(s)
Cardiomegaly/genetics , Hypertension/complications , Ventricular Dysfunction, Left/genetics , Animals , Disease Models, Animal , Disease Progression , Gene Expression Profiling/methods , Gene Expression Regulation , Genetic Markers , Hypertension/genetics , Male , Principal Component Analysis , RNA, Messenger/metabolism , Rats , Rats, Inbred Dahl , Reverse Transcriptase Polymerase Chain Reaction
7.
Hypertension ; 51(6): 1565-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18413490

ABSTRACT

Studies on genetically manipulated mice suggest a role for epsilon-protein kinase C (epsilonPKC) in cardiac hypertrophy and in heart failure. The potential clinical relevance of these findings was tested here using a pharmacological inhibitor of epsilonPKC activity during the progression to heart failure in hypertensive Dahl rats. Dahl rats, fed an 8% high-salt diet from the age of 6 weeks, exhibited compensatory cardiac hypertrophy by 11 weeks, followed by heart failure at approximately 17 weeks and death by the age of approximately 20 weeks (123+/-3 days). Sustained treatment between weeks 11 and 17 with the selective epsilonPKC inhibitor epsilonV1-2 or with an angiotensin II receptor blocker olmesartan prolonged animal survival by approximately 5 weeks (epsilonV1-2: 154+/-7 days; olmesartan: 149+/-5 days). These treatments resulted in improved fractional shortening (epsilonV1-2: 58+/-2%; olmesartan: 53+/-2%; saline: 41+/-6%) and decreased cardiac parenchymal fibrosis when measured at 17 weeks without lowering blood pressure at any time during the treatment. Combined treatment with epsilonV1-2, together with olmesartan, prolonged animal survival by 5 weeks (37 days) relative to olmesartan alone (from 160+/-5 to 197+/-14 days, respectively) and by approximately 11 weeks (74 days) on average relative to saline-treated animals, suggesting that the pathway inhibited by epsilonPKC inhibition is not identical to the olmesartan-induced effect. These data suggest that an epsilonPKC-selective inhibitor such as epsilonV1-2 may have a potential in augmenting current therapeutic strategies for the treatment of heart failure in humans.


Subject(s)
Heart Failure/drug therapy , Heart Failure/etiology , Hypertension/complications , Hypertension/drug therapy , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Cardiomegaly/drug therapy , Cardiomegaly/etiology , Cardiomegaly/pathology , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Heart Failure/pathology , Hypertension/pathology , Imidazoles/pharmacology , Male , Rats , Rats, Inbred Dahl , Sodium Chloride, Dietary/pharmacology , Tetrazoles/pharmacology
8.
Transplantation ; 85(6): 870-7, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18360270

ABSTRACT

BACKGROUND: This study aimed at investigating the efficacy of the novel immunosuppressant FK778 to prevent the development and progression of chronic allograft vasculopathy (CAV). METHODS: Orthotopic aortic transplantations were performed in the PVG-to-ACI rat model and followed over the course of 120 days. Immunosuppression with FK778 (20 mg/kg) or sirolimus (2 mg/kg) was either started early or delayed when CAV was already present. Trough levels were monitored. Aortic luminal obliteration was quantified using computer morphometry and intragraft cytokine profiles were analyzed with Western Blotting. Donor-reactive antibodies were quantified by flow cytometry. RESULTS: Untreated animals developed CAV with luminal obliteration of 25.2+/-13.6% and 41.4+/-23.3% after 80 and 120 days, respectively. Continuous immunosuppression with FK778 or sirolimus effectively prevented the development of vasculopathy. When the start of the immunosuppressive regimen was delayed until postoperative day 80, FK778 and sirolimus inhibited a progression of established CAV but did not reverse the luminal obliteration. Intragraft tumor growth factor-beta activity increased over the course of time in untreated recipients but was significantly suppressed after continuous immunosuppression with either agent. Expression of platelet-derived growth factor, intercellular adhesion molecule-1, and vascular adhesion molecule-1 also was moderately suppressed. A stable elevation of donor-reactive IgG-antibody levels was found over 120 days in the absence of treatment. With FK778 or sirolimus, antibody levels were effectively decreased. FK778 was very well tolerated and only sirolimus showed side effects with elevation of BUN, cholesterol, triglycerides, and ALT after 120 days. CONCLUSIONS: FK778 prevents the development of CAV and inhibits a progression of established disease. It shows a similar efficacy but a safer drug profile when compared to sirolimus.


Subject(s)
Alkynes/therapeutic use , Aorta/transplantation , Aortic Diseases/prevention & control , Isoxazoles/therapeutic use , Nitriles/therapeutic use , Transplantation, Homologous/pathology , Animals , Chronic Disease , Cyclosporine/therapeutic use , Immunosuppressive Agents/therapeutic use , Male , Rats , Rats, Inbred ACI
9.
Annu Rev Pharmacol Toxicol ; 48: 569-99, 2008.
Article in English | MEDLINE | ID: mdl-17919087

ABSTRACT

Cardiovascular disease is the leading cause of death in the United States. Therefore, identifying therapeutic targets is a major focus of current research. Protein kinase C (PKC), a family of serine/threonine kinases, has been identified as playing a role in many of the pathologies of heart disease. However, the lack of specific PKC regulators and the ubiquitous expression and normal physiological functions of the 11 PKC isozymes has made drug development a challenge. Here we discuss the validity of therapeutically targeting PKC, an intracellular signaling enzyme. We describe PKC structure, function, and distribution in the healthy and diseased heart, as well as the development of rationally designed isozyme-selective regulators of PKC functions. The review focuses on the roles of specific PKC isozymes in atherosclerosis, fibrosis, and cardiac hypertrophy, and examines principles of pharmacology as they pertain to regulators of signaling cascades associated with these diseases.


Subject(s)
Cardiovascular Diseases/drug therapy , Drug Delivery Systems , Protein Kinase C/drug effects , Animals , Cardiovascular Diseases/physiopathology , Chronic Disease , Drug Design , Humans , Isoenzymes/metabolism , Protein Kinase C/metabolism , Signal Transduction
10.
J Mol Cell Cardiol ; 43(4): 517-22, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17655859

ABSTRACT

Epsilon protein kinase C (epsilonPKC) plays pivotal roles in myocardial infarction and in heart failure. Although cardiac transplantation is a well-established therapy for severe heart failure, allograft rejection and host inflammatory responses limit graft function and reduce life expectancy. Here we determined whether sustained epsilonPKC inhibition beginning 3 days after transplantation suppress allograft rejection and improve cardiac transplantation using a murine heterotopic transplantation model. Hearts of FVB mice (H-2(q)) were transplanted into C57BL/6 mice (H-2(b)). Delivery of the epsilonPKC inhibitor, TAT(47-57)-epsilonV1-2 (epsilonV1-2, n=9, 20 mg/kg/day), or the carrier control peptide, TAT(47-57) (TAT, n=8), by osmotic pump began 3 days after transplantation and continued for the remaining 4 weeks. epsilonV1-2 treatment significantly improved the beating score throughout the treatment. Infiltration of macrophages and T cells into the cardiac grafts was significantly reduced and parenchymal fibrosis was decreased in animals treated with epsilonV1-2 as compared with control treatment. Finally, the rise in pro-fibrotic cytokine, TGF-beta and monocyte recruiting chemokine MCP-1 levels was almost abolished by epsilonV1-2 treatment, whereas the rise in PDGF-BB level was unaffected. These data suggest that epsilonPKC activity contributes to the chronic immune response in cardiac allograft and that an epsilonPKC-selective inhibitor, such as epsilonV1-2, could augment current therapeutic strategies to suppress inflammation and prolong graft survival in humans.


Subject(s)
Disease Models, Animal , Heart Failure/therapy , Heart Transplantation/adverse effects , Myocarditis/drug therapy , Myocarditis/etiology , Peptide Fragments/therapeutic use , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Animals , Chronic Disease , Cyclosporine/therapeutic use , Graft Rejection/etiology , Graft Survival/drug effects , Heart Failure/pathology , Heart Transplantation/pathology , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Peptide Fragments/pharmacology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes/drug effects , Transplantation, Heterotopic/adverse effects
11.
Biochem Biophys Res Commun ; 335(2): 550-7, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-16083853

ABSTRACT

RBCC protein interacting with PKC 1 (RBCK1) is a transcription factor belonging to the RING-IBR protein family and has been shown to shuttle between the nucleus and cytoplasm, possessing both the nuclear export and localization signals within its amino acid sequence. RBCK2, lacking the C-terminal half of RBCK1 including the RING-IBR domain, has also been identified as an alternative splice variant of RBCK1. RBCK2 shows no transcriptional activity and instead it represses the transcriptional activity of RBCK1. Here, we show that RBCK2 is present usually in the cytoplasm containing two Leu-rich regions that presumably serve as a nuclear export signal (NES). Moreover, an NES-disrupted RBCK1 that is mostly localized within the nucleus is translocated to the cytoplasm when coexpressed with RBCK2, suggesting that RBCK2 serves as a cytoplasmic tethering protein for RBCK1. We propose a novel and general function of RING-lacking splice variants of RING proteins to control the intracellular localization and functions of the parental RING proteins by forming a hetero-oligomeric complex.


Subject(s)
Cytoplasm/metabolism , Transcription Factors/chemistry , Alternative Splicing , Amino Acid Sequence , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Immunohistochemistry , Immunoprecipitation , Luciferases/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Plasmids/metabolism , Protein Binding , Protein Kinase C/metabolism , Protein Structure, Tertiary , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Ubiquitin-Protein Ligases
12.
J Biol Chem ; 280(24): 22937-44, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15833741

ABSTRACT

The intracellular localization of a RING-IBR protein, RBCK1, possessing DNA binding and transcriptional activities, has been investigated. The endogenous RBCK1 was found in both the cytoplasm and nucleus. Particularly in the nucleus, it was localized in the granular structures, most likely nuclear bodies. In contrast, the over-expressed RBCK1 was detected exclusively in the cytoplasm. When the cells were treated with leptomycin B, the over-expressed RBCK1 accumulated in the nuclear bodies. These results suggest that RBCK1 possesses the signal sequences responsible for the nuclearcytoplasmic translocation. Mutational analysis of RBCK1 has indicated that an N-terminal region containing Leu-142 and Leu-145 and a C-terminal one containing the RING-IBR domain serve as the nuclear export and localization signals, respectively. Thus, RBCK1 is a transcription factor dynamically shuttling between cytoplasm and nucleus. Furthermore, RBCK1 was found to interact with nuclear body proteins, CREB-binding protein (CBP), and promyelocytic leukemia protein (PML). Coexpression of RBCK1 with CBP significantly enhanced the transcriptional activity of RBCK1. Although PML per se showed no effect on the transcriptional activity of RBCK1, the CBP-enhanced activity was repressed by coexpression with PML, presumably through the interaction of PML and CBP. Taken together, our data demonstrate that RBCK1 is involved in transcriptional machinery in the nuclear bodies, and its transcriptional activity is regulated by nucleocytoplasmic shuttling.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/physiology , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , COS Cells , Cell Line , DNA Mutational Analysis , Down-Regulation , Genes, Reporter , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Leucine/chemistry , Luciferases/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Oligopeptides/chemistry , Plasmids/metabolism , Promyelocytic Leukemia Protein , Protein Structure, Tertiary , Protein Transport , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Tumor Suppressor Proteins , Up-Regulation
13.
Circulation ; 110(11 Suppl 1): II194-9, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15364862

ABSTRACT

BACKGROUND: Inhibiting delta protein kinase C (deltaPKC) during reperfusion and activating epsilon PKC (epsilonPKC) before ischemia each limits cardiac ischemic injury. Here, we examined whether limiting ischemia-reperfusion injury inhibits graft coronary artery disease (GCAD) and improves murine cardiac allografting. METHODS AND RESULTS: Hearts of FVB mice (H-2q) were transplanted into C57BL/6 mice (H-2b). epsilonPKC activator (psiepsilonRACK) was injected intraperitoneally (20 nmol) into donor mice 20 minutes before procurement. Hearts were then perfused with psiepsilonRACK (1.5 nmol) through the inferior vena cava (IVC) and subsequently submerged in psiepsilonRACK (0.5 micromol/L) for 20 minutes at 4 degrees C. Before reperfusion, the peritoneal cavity of recipients was irrigated with deltaPKC inhibitor (deltaV1-1, 300 nmol); control animals were treated with normal saline. The total ischemic time to the organ was 50 minutes. Two hours after transplantation, production of inflammatory cytokines and adhesion molecules, cardiomyocyte apoptosis, and caspase-3 and caspase-9 (but not caspase-8) activities were significantly reduced in the PKC regulator-treated group. Fas ligand levels (but not Fas) were also significantly reduced in this group. Importantly, GCAD indices, production of inflammatory cytokines, and adhesion molecules were significantly decreased and cardiac allograft function was significantly better as measured up to 30 days after transplantation. CONCLUSIONS: An epsilonPKC activator and a deltaPKC inhibitor together reduced GCAD. Clinically, these PKC isozyme regulators may be useful for organ preservation and prevention of ischemia-reperfusion injury and graft coronary artery disease in cardiac transplantation.


Subject(s)
Coronary Disease/drug therapy , Graft Rejection/drug therapy , Heart Transplantation , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/drug therapy , Oligopeptides/therapeutic use , Postoperative Complications/drug therapy , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Animals , Apoptosis/drug effects , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Adhesion Molecules/biosynthesis , Coronary Disease/etiology , Cytokines/biosynthesis , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Fas Ligand Protein , Graft Rejection/etiology , Inflammation/prevention & control , Injections, Intraperitoneal , Male , Membrane Glycoproteins/analysis , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Myocardial Ischemia/complications , Myocardial Reperfusion Injury/complications , Myocytes, Cardiac/pathology , Necrosis , Oligopeptides/administration & dosage , Peroxidase/analysis , Postoperative Complications/etiology , Protein Kinase C-delta , Protein Kinase C-epsilon , Therapeutic Irrigation , Transplantation, Heterotopic , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...