Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Pharmacol Toxicol Methods ; 64(3): 207-12, 2011.
Article in English | MEDLINE | ID: mdl-21843648

ABSTRACT

Antibody drug conjugates (ADCs) consist of an antibody attached to a cytotoxic drug by means of a linker. ADCs provide a way to couple the specificity of a monoclonal antibody (mAb) to the cytotoxicity of a small-molecule drug and, therefore, are promising new therapies for cancer. ADCs are prodrugs that are inactive in circulation but exert their cytotoxicity upon binding to the target cancer cell. Earlier unsuccessful attempts to generate ADCs with therapeutic value have emphasized the important role each component plays in determining the efficacy and safety of the final ADC. Scientific advances in engineering antibodies for maximum efficacy as anticancer agents, identification of highly cytotoxic molecules, and generation of linkers with increased stability in circulation have all contributed to the development of the many ADCs that are currently in clinical trials. This review discusses parameters that guide the selection of the components of an ADC to increase its therapeutic window, provides a brief look at ADCs currently in clinical trials, and discusses future challenges in this field.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Immunotoxins/immunology , Immunotoxins/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Humans
2.
Vet Pathol ; 48(3): 772-84, 2011 May.
Article in English | MEDLINE | ID: mdl-21383116

ABSTRACT

The pharmaceutical industry and regulatory agency toxicology testing paradigms in the United States currently appear successful, in part because of the continuously increasing life expectancy and the declining age-adjusted cancer rates in the United States. Although drugs likely have a minimal impact on the population statistics for cancer rates, pharmaceutical pathologists and toxicologists must focus on the individual risk for pharmaceutical carcinogenesis. As our understanding of carcinogenesis increases exponentially, and after hundreds if not thousands of rodent cancer tests, significant improvement in the precision of human pharmaceutical carcinogenesis hazard identification should now be possible and would enable a reduction in the substantial false-negative and false positive-rates reported herein. The appropriate use of acute, subchronic, chronic, and special toxicology tests to identify the major associated cancer risk factors, specifically, hormonal modulation, immunosuppression, genetic toxicity, and chronic toxicity, can be recognized through this review of pharmaceutical carcinogens. Significant opportunities exist for improving the effectiveness and efficiency of the current cancer risk assessment paradigm.


Subject(s)
Biological Assay/methods , Carcinogenicity Tests/methods , Drug-Related Side Effects and Adverse Reactions , Animals , Humans , Risk Factors , Rodentia
3.
Vet Pathol ; 47(2): 358-67, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20124009

ABSTRACT

Proteasome inhibitor therapeutics (PITs) have the potential to cause peripheral neuropathy. In a mouse model of PIT-induced peripheral neuropathy, the authors demonstrated that ubiquitin-positive multifocal protein aggregates with nuclear displacement appear in dorsal root ganglion cells of animals that subsequently develop nerve injuries. This peripheral-nerve effect in nonclinical models has generally been recognized as the correlate of grade 3 neuropathy in clinical testing. In differentiated PC12 cells, the authors demonstrated perturbations correlative with the development of neuropathy in vivo, including ubiquitinated protein aggregate (UPA) formation and/or nuclear displacement associated with the degree of proteasome inhibition. They compared 7 proteasome inhibitors of 3 chemical scaffolds (peptide boronate, peptide epoxyketone, and lactacystin analog) to determine if PIT-induced peripheral neuropathy is modulated by inhibition of the proteasome (ie, a mechanism-based effect) or due to effects independent of proteasome inhibition (ie, an off target or chemical-structure-based effect). The appearance of UPAs was assayed at IC(90) +/- 5% (90% inhibition concentration +/- 5%) for 20S proteasome inhibition. Results show that each of the investigated proteasome inhibitors induced identical proteasome-inhibitor-specific ubiquitin-positive immunostaining and nuclear displacement in PC12 cells. Other agents--such as paclitaxel, cisplatin, and thalidomide, which cause neuropathy by other mechanisms--did not cause UPAs or nuclear displacement, demonstrating that the effect was specific to proteasome inhibitors. In conclusion, PIT-induced neuronal cell UPA formation and nuclear displacement are mechanism based and independent of the proteasome inhibitor scaffold. These data indicate that attempts to modulate the neuropathy associated with PIT may not benefit from changing scaffolds.


Subject(s)
Peripheral Nervous System Diseases/chemically induced , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Ubiquitin/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , Cell Proliferation , Cell Survival , Dose-Response Relationship, Drug , Immunohistochemistry , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Oligopeptides/pharmacology , PC12 Cells , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System Diseases/metabolism , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , Rats , Ubiquitin/antagonists & inhibitors
4.
Toxicol Pathol ; 38(1): 148-64, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20075108

ABSTRACT

A daunting, unmet medical need exists for effective oncology chemotherapies, with cancer deaths in 2009 to exceed 560,000 in the United States alone. Because of the rapid demise of the majority of cancer patients with metastatic disease, oncology drug development must follow a much different paradigm than therapeutic candidates for less onerous diseases. The majority of drug candidates in development today are targeted at cancer therapy. Many of these candidate chemotherapeutic agents are active against novel targets, often presenting unique toxicological profiles. Since many of these novel targets are not unique to cancer cells, therapeutic margins may not exist. Decision making, in this event, is among the most challenging that any pharmaceutical toxicologist/pathologist or regulator will face. Nonclinical development scientists must compress timelines to present therapeutic options for cancer patients who have failed conventional therapy. In support of this goal, the U. S. Food and Drug Administration has created an oncology-specific paradigm for nonclinical testing and has introduced strategies to accelerate development and approval of successful candidates. Pharmaceutical toxicology testing strategies must not only satisfy regulation as the minimal expectation, but also attempt to reduce the current high attrition rates for oncologic candidates. A successful toxicology testing strategy represents the substance of this treatise.


Subject(s)
Antineoplastic Agents/adverse effects , Neoplasms/drug therapy , Toxicology/methods , Animals , Databases, Factual , Drug Discovery , Humans , Investigational New Drug Application , Maximum Tolerated Dose , Neoplasm Staging , Neoplasms/pathology
6.
Chem Biol Interact ; 150(1): 9-25, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15522258

ABSTRACT

The financial investment grows exponentially as a new chemical entity advances through each stage of discovery and development. The opportunity exists for the modern toxicologist to significantly impact expenditures by the early prediction of potential toxicity/side effect barriers to development by aggressive evaluation of development-limiting liabilities early in drug discovery. Improved efficiency in pharmaceutical research and development lies both in leveraging "best in class" technology and integration with pharmacologic activities during hit-to-lead and early lead optimization stages. To meet this challenge, a discovery assay by stage (DABS) paradigm should be adopted. The DABS clearly delineates to discovery project teams the timing and type of assay required for advancement of compounds to each subsequent level of discovery and development. An integrative core pathology function unifying Drug Safety Evaluation, Molecular Technologies and Clinical Research groups that effectively spans all phases of drug discovery and development is encouraged to drive the DABS. The ultimate goal of such improved efficiency being the accurate prediction of toxicity and side effects that would occur in development before commitment of the large prerequisite resource. Good justification of this approach is that every reduction of development attrition by 10% results in an estimated increase in net present value by $100 million.


Subject(s)
Toxicology/methods , Animals , Chemistry, Pharmaceutical , Drug Design , Drug Evaluation, Preclinical , Humans , Liver/pathology , Models, Animal , Reproducibility of Results , Research Design
7.
Biochem Biophys Res Commun ; 283(5): 1061-8, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11355880

ABSTRACT

Action of protein kinases and phosphatases contributes to myocardial hypertrophy. PRL-3, a protein tyrosine phosphatase, was identified in a cDNA library from an explanted human heart obtained from a patient with idiopathic cardiomyopathy. PRL-3 is expressed in heart and skeletal muscle, exhibiting approximately 76% identity to the ubiquitous tyrosine phosphatase PRL-1, which was reported to increase cell proliferation. PRL-3 was cloned into E. coli and purified using affinity chromatography. PRL-3 activity was determined using the substrate 6,8-difluoro-4-methylumbelliferyl phosphate, and was inhibited by vanadate and analogs. HEK293 cells expressing PRL-3 demonstrated increased growth rates versus nontransfected cells or cells transfected with the catalytically inactive C104S PRL-3 mutant. The tyrosine phosphatase inhibitor, potassium bisperoxo (bipyridine) oxovanadate V, normalizes the growth rate of PRL-3 expressing cells to that of parental HEK293 cells in a concentration-dependent manner. Using FLIPR analysis, parental HEK293 cells mobilize calcium when stimulated with angiotensin-II (AngII). However, calcium mobilization is inhibited in cells expressing wild-type PRL-3 when stimulated with AngII, while cells expressing the inactive mutant of PRL-3 mobilize calcium to the same extent as parental HEK293 cells. Western blots comparing PRL-3 transfected cells to parental HEK293 cells showed dephosphorylation of p130(cas) in response to AngII. These data suggest a role for PRL-3 in the modulation of intracellular calcium transients induced by AngII.


Subject(s)
Angiotensin II/pharmacology , Calcium Signaling/physiology , Calcium/metabolism , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Muscle, Skeletal/enzymology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Signal Transduction/physiology , Amino Acid Substitution , Calcium Signaling/drug effects , Cardiomyopathies/enzymology , Cardiomyopathies/genetics , Cell Division/drug effects , Cell Line , Chromatography, Affinity , Cloning, Molecular , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Escherichia coli , Gene Library , Humans , Immediate-Early Proteins/isolation & purification , Mutagenesis, Site-Directed , Myocardium/enzymology , Neoplasm Proteins , Organ Culture Techniques , Organometallic Compounds/pharmacology , Phenanthrolines/pharmacology , Protein Tyrosine Phosphatases/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection , Vanadates/pharmacology
8.
Circulation ; 103(6): 889-96, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11171800

ABSTRACT

BACKGROUND: Relieving the inhibition of sarcoplasmic reticular function by phospholamban is a major target of beta-adrenergic stimulation. Chronic beta-adrenergic receptor activity has been suggested to be detrimental, on the basis of transgenic overexpression of the receptor or its signaling effectors. However, it is not known whether physiological levels of sympathetic tone, in the absence of preexisting heart failure, are similarly detrimental. METHODS AND RESULTS: Transgenic mice overexpressing phospholamban at 4-fold normal levels were generated, and at 3 months, they exhibited mildly depressed ventricular contractility without heart failure. As expected, transgenic cardiomyocyte mechanics and calcium kinetics were depressed, but isoproterenol reversed the inhibitory effects of phospholamban on these parameters. In vivo cardiac function was substantially depressed by propranolol administration, suggesting enhanced sympathetic tone. Indeed, plasma norepinephrine levels and the phosphorylation status of phospholamban were elevated, reflecting increased adrenergic drive in transgenic hearts. On aging, the chronic enhancement of adrenergic tone was associated with a desensitization of adenylyl cyclase (which intensified the inhibitory effects of phospholamban), the development of overt heart failure, and a premature mortality. CONCLUSIONS: The unique interaction between phospholamban and increased adrenergic drive, elucidated herein, provides the first evidence that compensatory increases in catecholamine stimulation can, even in the absence of preexisting heart failure, be a primary causative factor in the development of cardiomyopathy and early mortality.


Subject(s)
Aging , Calcium-Binding Proteins/metabolism , Cardiomyopathies/etiology , Receptors, Adrenergic, beta/metabolism , Adenylyl Cyclases/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Cardiomyopathies/blood , Cardiomyopathies/mortality , Echocardiography , Heart Failure/etiology , Heart Failure/mortality , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Norepinephrine/blood , Phosphorylation , Propranolol/pharmacology , Ventricular Function, Left
9.
J Mol Cell Cardiol ; 32(9): 1735-44, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10966834

ABSTRACT

Cardiac-specific overexpression of calsequestrin has been shown to result in significant decreases in contractile parameters and intracellular Ca(2+)transients in vitro. Therefore, the purpose of the present study was to determine the effects of calsequestrin overexpression on basal cardiac function and the force-frequency relation in vivo. Calsequestrin overexpression mice (CSQ-OE, n=20) and their isogenic controls (WT) were studied with an integrative approach using transthoracic echocardiography, stress-shortening relations, and invasive hemodynamics in intact closed-chest mice. M-mode echocardiography indicated that calsequestrin overexpression resulted in concentric hypertrophy (+52%) and an increase in LV ejection phase indices. However, mean end-systolic stress-shortening coordinates revealed that at matched end-systolic wall-stress, fractional shortening was depressed in CSQ-OE mice. This was confirmed by depressed indices of LV isovolumic contraction and relaxation in CSQ-OE v. WT mice. Furthermore, overexpression of calsequestrin resulted in a downward and leftward shift of the biphasic force-frequency relation; thus, the critical heart (HR(crit)) was significantly lower in calsequestrin-overexpression mice (264+/-15 bpm) than in wild-type controls (365+/-21 bpm). Surprisingly, calsequestrin overexpression was associated with the induction of pulsus alternans in every animal (at an average heart rate of 428+/-26 bpm), whereas none of the wild-type controls displayed this phenomenon. We conclude that: (i) although increased levels of calsequestrin result in decreased myocardial contractility and a depressed force-frequency relation, LV wall stress is reduced and chamber function is normal, and (ii) an increase in SR Ca(2+)storage capacity induces pulsus alternans in the intact anesthetized mouse.


Subject(s)
Calsequestrin/physiology , Hypertrophy, Left Ventricular/etiology , Animals , Echocardiography , Gene Expression , Hemodynamics , Hypertrophy, Left Ventricular/physiopathology , Mice , Mice, Transgenic , Myocardial Contraction
10.
Am J Physiol Heart Circ Physiol ; 278(3): H958-63, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10710365

ABSTRACT

Mechanical and relaxation restitution represent the restoration of contractile force and relaxation, respectively, in premature beats having progressively longer extrasystolic intervals (ESI); these phenomena are related to intracellular activator Ca(2+) by poorly defined mechanisms. We tested the hypothesis that the level of phospholamban [which modulates the affinity of the sarcoplasmic reticulum (SR) Ca(2+)-ATPase for Ca(2+), and thus the SR Ca(2+) load] may be an important determinant of both mechanical and relaxation restitution. Five mice with ablation of the phospholamban (PLB) gene (PLBKO), eight isogenic wild-type controls (129SvJ), eleven mice with PLB overexpression (PLBOE), and nine isogenic wild-type (FVB/N) controls were anesthetized and instrumented with a 1.4-Fr Millar catheter in the left ventricle and a 1-Fr pacemaker in the right atrium. At a cycle length of 200 ms, extrastimuli with increasing ESI were introduced, and the peak rates of left ventricular isovolumic contraction (+/-dP/dt(max)) were normalized and fit to monoexponential equations. In a subset, the protocols were repeated after ryanodine (4 ng/g) was administered to deplete SR Ca(2+) stores. The time constant of mechanical restitution in PLBKO was significantly shorter [6.3 +/- 1.2 (SE) vs. 47.7 +/- 7.6 ms] and began earlier (50 +/- 10 vs. 70 +/- 19 ms) than in 129SvJ. In contrast, the time constant of mechnical restitution was significantly longer (80.3 +/- 7.6 vs. 54.1 +/- 9.2 ms) in PLBOE than in FVB/N. The time constant of relaxation restitution was less in PLBKO than in 129SvJ (26.2 +/- 9.9 vs. 44.6 +/- 3.3, P < 0.05) but was similar in PLBOE and FVB/N (21.1 +/- 6.3 vs. 20.5 +/- 5.7 ms). Intravenous ryanodine decreased significantly the time constants of mechanical restitution in PLBOE, 129SvJ, and FVB/N but was lethal in PLBKO. In contrast, ryanodine increased the time constant of relaxation restitution. Thus 1) the phospholamban level is a critical determinant of mechanical restitution and (to a lesser extent) relaxation restitution in these transgenic models, and 2) ryanodine differentially affects mechanical and relaxation restitution. Furthermore, our data suggest a dissociation of processes within the SR that govern contraction and relaxation.


Subject(s)
Calcium/metabolism , Myocardial Contraction/physiology , Sarcoplasmic Reticulum/physiology , Animals , Biomechanical Phenomena , Blood Pressure , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Heart Rate , Mice , Mice, Knockout , Ryanodine/pharmacology , Sarcoplasmic Reticulum/drug effects
11.
J Mol Cell Cardiol ; 31(11): 2007-15, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10591027

ABSTRACT

Twelve mice with PLB overexpression (PLBOE), and 11 isogenic FVB/N wild-type (WT) controls, were anesthetized and instrumented with a 1.4 F Millar catheter in the LV and a 1 F pacemaker in the right atrium. At a cycle length of 200 ms and a fixed extrastimulus of 120 ms, extrastimuli with increasing intervals (PESI) up to 1000 ms were introduced, and the peak rates of LV isovolumic contraction (+/- dP/dtmax) were normalized and fit to monoexponential equations. In a subset of animals, the protocols were repeated after ryanodine (4 ng/g) was given to deplete SR Ca2+ stores. The time constant and the plateau of the exponential curve fits were significantly greater in PLBOE than WT (107.8 +/- 7.0 v 75.2 +/- 5.5 ms and 1.39 +/- 0.03 v 1.08 +/- 0.02, both P < 0.05). At 200, 600 and 1000 ms, the normalized dP/dt was significantly greater in PLBOE than WT. After ryanodine, normalized dP/dt was significantly decreased in PLBOE, but unchanged in WT. The protein levels of the sodium-calcium exchanger normalized to calsequestrin were increased 3.7 +/- 0.3-fold in PLBOE compared to controls. In conclusion, the phospholamban level is a critical determinant of postextrasystolic potentiation in this transgenic model, and is differentially impaired by ryanodine at long diastolic intervals in PLBOE v controls. These differences may be due in part to changes in the protein level and resultant activity of the sodium calcium exchanger.


Subject(s)
Calcium-Binding Proteins/physiology , Cardiac Complexes, Premature/physiopathology , Hemodynamics , Adenosine Triphosphatases/metabolism , Animals , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cardiac Complexes, Premature/genetics , Diastole , Electric Stimulation , Electrocardiography , Female , Heart/drug effects , Heart/physiology , Heart/physiopathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Ryanodine/pharmacology , Systole , Ventricular Function, Left
12.
Am J Physiol ; 276(6): H2245-50, 1999 06.
Article in English | MEDLINE | ID: mdl-10362708

ABSTRACT

Phospholamban levels regulate cardiac sarcoplasmic reticulum Ca2+ pump activity and myocardial contractility. To determine whether and to what extent phospholamban modulates the force-frequency relation and ventricular relaxation in vivo, we studied transgenic mice overexpressing phospholamban (PLBOE), gene-targeted mice without phospholamban (PLBKO), and isogenic wild-type controls. Contractility was assessed by the peak rate of left ventricular (LV) isovolumic contraction (+dP/dtmax), and diastolic function was assessed by both the peak rate (-dP/dtmax) and the time constant (tau) of isovolumic LV relaxation, using a high-fidelity LV catheter. Incremental atrial pacing was used to generate heart rate vs. -dP/dtmax (force-frequency) relations. Biphasic force-frequency relations were produced in all animals, and the critical heart rate (HRcrit) was taken as the heart rate at which dP/dtmax was maximal. The average LV +dP/dtmax increased in both PLBKO and PLBOE compared with their isogenic controls (both P < 0.05). The HRcrit for LV +dP/dtmax was significantly higher in PLBKO (427 +/- 20 beats/min) compared with controls (360 +/- 18 beats/min), whereas the HRcrit in PLBOE (340 +/- 30 beats/min) was significantly lower compared with that in isogenic controls (440 +/- 25 beats/min). The intrinsic heart rates were significantly lower, and the HRcrit and the +/-dP/dtmax at HRcrit were significantly greater in FVB/N than in SvJ control mice. We conclude that 1) the level of phospholamban is a critical negative determinant of the force-frequency relation and myocardial contractility in vivo, and 2) contractile parameters may differ significantly between strains of normal mice.


Subject(s)
Calcium-Binding Proteins/physiology , Heart Rate/physiology , Myocardial Contraction/physiology , Animals , Calcium-Binding Proteins/genetics , Female , Hemodynamics/physiology , Male , Mice , Mice, Inbred Strains , Mice, Knockout/genetics , Species Specificity
13.
Circ Res ; 84(3): 360-4, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-10024311

ABSTRACT

Vascular endothelial cells regulate vascular smooth muscle tone through Ca2+-dependent production and release of vasoactive molecules. Phospholamban (PLB) is a 24- to 27-kDa phosphoprotein that modulates activity of the sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA). Expression of PLB is reportedly limited to cardiac, slow-twitch skeletal and smooth muscle in which PLB is an important regulator of [Ca2+]i and contractility in these muscles. In the present study, we report the existence of PLB in the vascular endothelium, a nonmuscle tissue, and provide functional data on PLB regulation of vascular contractility through its actions in the endothelium. Endothelium-dependent relaxation to acetylcholine was attenuated in aorta of PLB-deficient (PLB-KO) mice compared with wild-type (WT) controls. This effect was not due to actions of nitric oxide on the smooth muscle, because sodium nitroprusside-mediated relaxation in either denuded or endothelium-intact aortas was unaffected by PLB ablation. Relative to denuded vessels, relaxation to forskolin was enhanced in WT endothelium-intact aortas. The endothelium-dependent component of this relaxation was attenuated in PLB-KO aortas. To investigate whether these changes were due to PLB, WT mouse aorta endothelial cells were isolated. Both reverse transcriptase-polymerase chain reaction and Western blot analyses revealed the presence of PLB in endothelial cells, which were shown to be >98% pure by diI-acetylated LDL uptake and nuclear counterstaining. These data indicate that PLB is present and modulates vascular function as a result of its actions in endothelial cells. The presence of PLB in endothelial cells opens new fields for investigation of Ca2+ regulatory pathways in nonmuscle cells and for modulation of endothelial-vascular interactions.


Subject(s)
Calcium-Binding Proteins/physiology , Endothelium, Vascular/physiology , Vasodilation , Animals , Calcium/metabolism , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/genetics , Cells, Cultured , Colforsin/pharmacology , Endothelium, Vascular/chemistry , Mice , Mice, Knockout , Nitroprusside/pharmacology , Vasodilation/drug effects
14.
J Biol Chem ; 273(50): 33674-80, 1998 Dec 11.
Article in English | MEDLINE | ID: mdl-9837953

ABSTRACT

Phospholamban has been proposed to coexist as pentamers and monomers in native sarcoplasmic reticulum membranes. To determine its functional unit in vivo, we reintroduced wild-type (pentameric) or monomeric mutant (C41F) phospholamban in the hearts of phospholamban knockout mice. Transgenic lines, expressing similar levels of mutant or wild-type phospholamban, were identified, and their cardiac phenotypes were characterized in parallel. Sarcoplasmic reticulum Ca2+ transport assays indicated similar decreases in SERCA2 Ca2+ affinity by mutant or wild-type phospholamban. However, the time constants of relaxation and Ca2+ transient decline in isolated cardiomyocytes were diminished to a greater extent by wild-type than mutant phospholamban, even without significant differences in the amplitudes of myocyte contraction and Ca2+ transients between the two groups. Langendorff perfusion also indicated that mutant phospholamban was not capable of depressing the enhanced relaxation parameters of the phospholamban knockout hearts to the same extent as wild-type phospholamban. Moreover, in vivo assessment of mouse hemodynamics revealed a greater depression of cardiac function in wild-type than mutant phospholamban hearts. Thus, the mutant or monomeric form of phospholamban was not as effective in slowing Ca2+ decline or relaxation in cardiomyocytes, hearts, or intact animals as wild-type or pentameric phospholamban. These findings suggest that pentameric assembly of phospholamban is necessary for optimal regulation of myocardial contractility in vivo.


Subject(s)
Calcium-Binding Proteins/metabolism , Heart/physiology , Myocardium/metabolism , Animals , Biopolymers , Calcium/metabolism , Calcium-Binding Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mutagenesis, Site-Directed , Myocardial Contraction , Phosphorylation , Sarcoplasmic Reticulum/metabolism
15.
J Biol Chem ; 273(43): 28470-7, 1998 Oct 23.
Article in English | MEDLINE | ID: mdl-9774476

ABSTRACT

Calsequestrin is a high capacity Ca2+-binding protein in the sarcoplasmic reticulum (SR) lumen. To elucidate the functional role of calsequestrin in vivo, transgenic mice were generated that overexpressed mouse cardiac calsequestrin in the heart. Overexpression (20-fold) of calsequestrin was associated with cardiac hypertrophy and induction of a fetal gene expression program. Isolated transgenic cardiomyocytes exhibited diminished shortening fraction (46%), shortening rate (60%), and relengthening rate (60%). The Ca2+ transient amplitude was also depressed (45%), although the SR Ca2+ storage capacity was augmented, as suggested by caffeine application studies. These alterations were associated with a decrease in L-type Ca2+ current density and prolongation of this channel's inactivation kinetics without changes in Na+-Ca2+ exchanger current density. Furthermore, there were increases in protein levels of SR Ca2+-ATPase, phospholamban, and calreticulin and decreases in FKBP12, without alterations in ryanodine receptor, junctin, and triadin levels in transgenic hearts. Left ventricular function analysis in Langendorff perfused hearts and closed-chest anesthetized mice also indicated depressed rates of contraction and relaxation of transgenic hearts. These findings suggest that calsequestrin overexpression is associated with increases in SR Ca2+ capacity, but decreases in Ca2+-induced SR Ca2+ release, leading to depressed contractility in the mammalian heart.


Subject(s)
Calsequestrin/metabolism , Cardiomegaly , Myocardial Contraction , Ventricular Function, Left , Amino Acid Sequence , Animals , Base Sequence , Calcium Channels/metabolism , Calsequestrin/genetics , Cells, Cultured , DNA, Complementary/genetics , Gene Expression , Gene Expression Regulation, Developmental , Gene Library , In Vitro Techniques , Mice , Mice, Transgenic , Molecular Sequence Data , Myocardium/cytology , Myocardium/ultrastructure , Perfusion , Recombinant Proteins/metabolism , Sarcoplasmic Reticulum/metabolism , Sodium-Calcium Exchanger/metabolism
16.
J Mol Cell Cardiol ; 30(7): 1275-84, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9710796

ABSTRACT

Phospholamban gene transcript levels are much lower in murine atria as compared to murine ventricles and this reduced phospholamban expression has been suggested to result in enhanced atrial contractile parameters. To delineate the functional role of phospholamban in murine atrium, the contractile parameters of isolated muscles from phospholamban knockout and cardiac-specific phospholamban overexpression mice along with their isogenic wild-type controls were evaluated. Assessment of the times (ms) to peak tension development and to half-relaxation of developed tension, as well as the rates (mg/s) of tension development and relaxation in paced atrial muscles, revealed that phospholamban ablation was associated with enhanced rates of relaxation with no significant effect on contraction rate, while phospholamban overexpression (three-fold) was associated with depressed rates of both contraction and relaxation. Isoproterenol stimulation resulted in significant increases in the rates of developed tension and relaxation in both phospholamban deficient and phospholamban overexpression atria, indicating that the beta-adrenergic pathway was functional in these muscles. These findings suggest that phospholamban is an important modulator of atrial contractility and its responses to beta-adrenergic agonists.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Atrial Function , Calcium-Binding Proteins/physiology , Isoproterenol/pharmacology , Myocardial Contraction/physiology , Animals , Calcium-Binding Proteins/genetics , Female , Heart Atria/drug effects , Mice , Mice, Knockout , Myocardial Contraction/drug effects
17.
J Mol Cell Cardiol ; 30(2): 317-25, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9515008

ABSTRACT

In mammalian ventricular myocytes, inactivation of L-type Ca2+ channels (CaCh) is controlled by voltage- and Ca2+-dependent mechanisms. The Ca2+-dependent component is regulated by the Ca2+ released from the sarcoplasmic reticulum (SR). However, little is known about the inactivation properties of CaCh in atrial myocytes, which lack spatial coupling between CaCh and SR Ca2+ release channels. The cardiac SR Ca2+ load is determined by the activity of SR Ca2+-ATPase, which is inversely regulated by the levels of phospholamban (PLB). To investigate the role of SR Ca2+ in atrial myocytes, Ca2+ currents (I Ca) were recorded in mouse atrial myocytes recorded from wild-type (WT) mice and the characteristics were compared to those obtained from atrial myocytes from the transgenic mice overexpressing PLB (PLB-OEX). ICa from WT exhibited fast and slow components of inactivation and the rate of inactivation was slowed when SR Ca2+ was depleted by caffeine, suggesting that the inactivation of atrial ICa is modulated by SR Ca2+ load. The current density and voltage-dependence of ICa were similar between the two groups. However, the fast component of inactivation was significantly reduced in PLB-OEX. When Ca2+ was replaced by Ba2+ or in the presence of caffeine, inactivation was slowed and the decay of the current was not significantly different between WT and PLB-OEX. These results suggest that the inactivation of ICa in mouse atrial myocytes involves Ca2+-dependent and voltage-dependent components. The decrease in the faster component of inactivation in PLB-OEX is consistent with the idea that CaCh and SR Ca2+ release channels are functionally coupled and Ca2+ released from the SR contributes the Ca2+-dependent inactivation component.


Subject(s)
Calcium Channels/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Myocardium/metabolism , Animals , Calcium Channels, L-Type , Gene Expression , Heart Atria/cytology , Heart Atria/metabolism , In Vitro Techniques , Kinetics , Membrane Potentials , Mice , Mice, Transgenic , Myocardium/cytology , Patch-Clamp Techniques , Sarcoplasmic Reticulum/metabolism
18.
J Biol Chem ; 273(8): 4734-9, 1998 Feb 20.
Article in English | MEDLINE | ID: mdl-9468536

ABSTRACT

Phospholamban is a critical regulator of the sarcoplasmic reticulum Ca2+-ATPase activity and myocardial contractility. Phosphorylation of phospholamban occurs on both Ser16 and Thr17 during isoproterenol stimulation. To determine the physiological significance of dual site phospholamban phosphorylation, we generated transgenic models expressing either wild-type or the Ser16 --> Ala mutant phospholamban in the cardiac compartment of the phospholamban knockout mice. Transgenic lines with similar levels of mutant or wild-type phospholamban were studied in parallel. Langendorff perfusion indicated that the basal hyperdynamic cardiac function of the knockout mouse was reversed to the same extent by reinsertion of either wild-type or mutant phospholamban. However, isoproterenol stimulation was associated with much lower responses in the contractile parameters of mutant phospholamban compared with wild-type hearts. These attenuated responses were due to lack of phosphorylation of mutant phospholamban, assessed in 32P labeling perfusion experiments. The lack of phospholamban phosphorylation in vivo was not due to conversion of Ser16 to Ala, since the mutated phospholamban form could serve as substrate for the calcium-calmodulin-dependent protein kinase in vitro. These findings indicate that phosphorylation of Ser16 is a prerequisite for Thr17 phosphorylation in phospholamban, and prevention of phosphoserine formation results in attenuation of the beta-agonist stimulatory responses in the mammalian heart.


Subject(s)
Calcium-Binding Proteins/metabolism , Animals , Calcium-Binding Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mutagenesis, Site-Directed , Myocardium/metabolism , Phosphorus Radioisotopes/metabolism , Phosphorylation
20.
Biochem Biophys Res Commun ; 239(1): 1-5, 1997 Oct 09.
Article in English | MEDLINE | ID: mdl-9345259

ABSTRACT

Phospholamban is a major regulator of the kinetics of cardiac contractility, through its ability to regulate the function of the cardiac SR Ca(2+)-pump and thus the SR Ca2+ load. In vitro expression studies have provided significant information on the structure/function of the phospholamban/Ca(2+)-pump interaction. Furthermore, the generation of genetic animal models with altered phospholamban expression levels have permitted a through understanding of the physiological role of this regulatory phosphoprotein. Future studies aimed towards crystallization of phospholamban and the SR Ca(2+)-ATPase in their native SR environment may provide clues to their tertiary and quaternary structures and may further elucidate the mechanisms underlying the phospholamban regulatory effects in vivo.


Subject(s)
Calcium-Binding Proteins/physiology , Calcium-Transporting ATPases/physiology , Animals , Calcium-Binding Proteins/genetics , Cloning, Molecular , Humans , Models, Molecular , Myocardial Contraction/physiology , Myocardium/enzymology , Sarcoplasmic Reticulum/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...