Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ESC Heart Fail ; 10(1): 465-477, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36300679

ABSTRACT

AIMS: Peripartum cardiomyopathy (PPCM) is a rare heart disease, occurring in previously heart-healthy women during the last month of pregnancy or the first months after delivery due to left ventricular (LV) systolic dysfunction. A common pathomechanistic pathway of PPCM includes increased oxidative stress and the subsequent generation of a cleaved prolactin fragment (16 kDa PRL), which promotes the onset of heart failure (HF) in a microRNA (miR)-146a-dependent manner. Inhibition of prolactin secretion with the dopamine D2 receptor (D2R) agonist bromocriptine combined with standard HF therapy supports cardiac recovery. This study examined whether treatment with the more selective D2R agonist cabergoline prevents HF development in an experimental PPCM mouse model and might be used as an alternative treatment regime for PPCM. METHODS AND RESULTS: Postpartum (PP) female PPCM-prone mice with a cardiomyocyte restricted STAT3-deficiency (αMHC-Cretg/+ ; Stat3fl/fl ; CKO) were treated over two consecutive nursing periods with cabergoline (CKO Cab, 0.5 mg/kg/day) and were compared with bromocriptine treated CKO (CKO Br) and postpartum-matched WT and CKO mice. Cabergoline treatment in CKO PP mice preserved cardiac function [fractional shortening (FS): CKO Cab: 34.5 ± 9.4% vs. CKO: 22.1 ± 9%, P < 0.05] and prevented the development of cardiac hypertrophy, fibrosis, and inflammation as effective as bromocriptine therapy (FS: CKO Br: 33.4 ± 5.6%). The myocardial up-regulation of the PPCM biomarkers plasminogen inhibitor activator 1 (PAI-1) and miR-146a were prevented by both cabergoline and bromocriptine therapy. A small cohort of three PPCM patients from the German PPCM Registry was treated with cabergoline (1 mg per week for 2 weeks, followed by 0.5 mg per week for another 6 weeks) due to a temporary unavailability of bromocriptine. All PPCM patients initially presented with a severely reduced LV ejection fraction (LVEF: 26 ± 2%). However, at 6 months of follow-up, LV function (LVEF: 56 ± 2%) fully recovered in all three PPCM patients, and no adverse events were detected. CONCLUSIONS: In the experimental PPCM mouse model, the selective D2R agonist cabergoline prevents the onset of postpartum HF similar to bromocriptine. In PPCM patients, cabergoline treatment was safe and effective as all patients fully recovered. Cabergoline might serve as a promising alternative to bromocriptine. However, these findings are based on experimental data and a small case series and thus have to be interpreted with caution and should be validated in a larger clinical trial.


Subject(s)
Cardiomyopathies , Heart Failure , MicroRNAs , Ventricular Dysfunction, Left , Pregnancy , Female , Mice , Animals , Bromocriptine , Cabergoline/metabolism , Cabergoline/therapeutic use , Peripartum Period , Prolactin/metabolism , Prolactin/therapeutic use , Heart Failure/drug therapy , Myocytes, Cardiac/metabolism , Dopamine Agonists , Ventricular Dysfunction, Left/drug therapy , MicroRNAs/metabolism
2.
Am J Physiol Heart Circ Physiol ; 323(6): H1352-H1364, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36399384

ABSTRACT

Perturbed vitamin-A metabolism is associated with type 2 diabetes and mitochondrial dysfunction that are pathophysiologically linked to the development of diabetic cardiomyopathy (DCM). However, the mechanism, by which vitamin A might regulate mitochondrial energetics in DCM has previously not been explored. To test the hypothesis that vitamin-A deficiency accelerates the onset of cardiomyopathy in diet-induced obesity (DIO), we subjected mice with lecithin retinol acyltransferase (Lrat) germline deletion, which exhibit impaired vitamin-A stores, to vitamin A-deficient high-fat diet (HFD) feeding. Wild-type mice fed with a vitamin A-sufficient HFD served as controls. Cardiac structure, contractile function, and mitochondrial respiratory capacity were preserved despite vitamin-A deficiency following 20 wk of HFD feeding. Gene profiling by RNA sequencing revealed that vitamin A is required for the expression of genes involved in cardiac fatty acid oxidation, glycolysis, tricarboxylic acid cycle, and mitochondrial oxidative phosphorylation in DIO as expression of these genes was relatively preserved under vitamin A-sufficient HFD conditions. Together, these data identify a transcriptional program, by which vitamin A preserves cardiac energetic gene expression in DIO that might attenuate subsequent onset of mitochondrial and contractile dysfunction.NEW & NOTEWORTHY The relationship between vitamin-A status and the pathogenesis of diabetic cardiomyopathy has not been studied in detail. We assessed cardiac mitochondrial respiratory capacity, contractile function, and gene expression by RNA sequencing in a murine model of combined vitamin-A deficiency and diet-induced obesity. Our study identifies a role for vitamin A in preserving cardiac energetic gene expression that might attenuate subsequent development of mitochondrial and contractile dysfunction in diet-induced obesity.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Mice , Animals , Vitamin A , Disease Models, Animal , Diet , Obesity/genetics , Gene Expression , Vitamins
3.
Cancers (Basel) ; 14(4)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35205731

ABSTRACT

Targeted therapies are currently considered the best cost-benefit anti-cancer treatment. In hematological malignancies, however, relapse rates and non-hematopoietic side effects including cardiotoxicity remain high. Here, we describe significant heart damage due to advanced acute lymphoblastic leukemia (ALL) with t(9;22) encoding the bcr-abl oncogene (BCR-ABL+ ALL) in murine xenotransplantation models. Echocardiography reveals severe cardiac dysfunction with impaired left ventricular function and reduced heart and cardiomyocyte dimensions associated with increased apoptosis. This cardiac damage is fully reversible, but cardiac recovery depends on the therapy used to induce ALL remission. Chemotherapy-free combination therapy with dasatinib (DAS), venetoclax (VEN) (targeting the BCR-ABL oncoprotein and mitochondrial B-cell CLL/Lymphoma 2 (BCL2), respectively), and dexamethasone (DEX) can fully revert cardiac defects, whereas the depletion of otherwise identical ALL in a genetic model using herpes simplex virus type 1 thymidine kinase (HSV-TK) cannot. Mechanistically, dexamethasone induces a pro-apoptotic BCL2-interacting mediator of cell death (BIM) expression and apoptosis in ALL cells but enhances pro-survival B-cell lymphoma extra-large (BCLXL) expression in cardiomyocytes and clinical recovery with the reversion of cardiac atrophy. These data demonstrate that therapies designed to optimize apoptosis induction in ALL may circumvent cardiac on-target side effects and may even activate cardiac recovery. In the future, combining the careful clinical monitoring of cardiotoxicity in leukemic patients with the further characterization of organ-specific side effects and signaling pathways activated by malignancy and/or anti-tumor therapies seems reasonable.

4.
Blood ; 137(19): 2657-2661, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33512436

ABSTRACT

Adult patients with relapsed B-cell precursor acute lymphoblastic leukemia (BCP-ALL) have a dismal prognosis. To improve pharmacotherapy, we analyzed induction of apoptosis by venetoclax and inotuzumab ozogamicin in terms of cytotoxicity and mode of action. Flow cytometry-based analyses of mitochondrial outer membrane permeabilization (MOMP) and ataxia telangiectasia mutated activation demonstrate rapid induction of MOMP by venetoclax and DNA damage signaling by inotuzumab ozogamicin, respectively. In primary ALL samples and patient-derived xenograft (PDX) models, venetoclax and inotuzumab ozogamicin cooperated and synergized in combination with dexamethasone in vitro in all tested samples of ALL. In murine PDX models, inotuzumab ozogamicin, but not venetoclax, induced complete remission in a dose-dependent manner but constantly failed to achieve relapse-free survival. In contrast, combination therapy with venetoclax, dexamethasone, and inotuzumab ozogamicin induced long-term leukemia-free survival and treatment-free survival in all 3 ALL-PDX models tested. These data demonstrate synergistic and highly efficient pharmacotherapy in preclinical models that qualify for evaluation in clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , DNA Damage , DNA, Neoplasm/drug effects , Dexamethasone/pharmacology , Inotuzumab Ozogamicin/pharmacology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Sulfonamides/pharmacology , Adolescent , Adult , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Calicheamicins/pharmacology , DNA Breaks, Double-Stranded , Dexamethasone/administration & dosage , Drug Synergism , Female , Humans , Inotuzumab Ozogamicin/administration & dosage , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Mitochondrial Membranes/drug effects , Recurrence , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays
5.
PLoS Biol ; 18(12): e3000739, 2020 12.
Article in English | MEDLINE | ID: mdl-33370269

ABSTRACT

Cardiac levels of the signal transducer and activator of transcription factor-3 (STAT3) decline with age, and male but not female mice with a cardiomyocyte-specific STAT3 deficiency conditional knockout (CKO) display premature age-related heart failure associated with reduced cardiac capillary density. In the present study, isolated male and female CKO-cardiomyocytes exhibit increased prostaglandin (PG)-generating cyclooxygenase-2 (COX-2) expression. The PG-degrading hydroxyprostaglandin-dehydrogenase-15 (HPGD) expression is only reduced in male cardiomyocytes, which is associated with increased prostaglandin D2 (PGD2) secretion from isolated male but not female CKO-cardiomyocytes. Reduced HPGD expression in male cardiomyocytes derive from impaired androgen receptor (AR)-signaling due to loss of its cofactor STAT3. Elevated PGD2 secretion in males is associated with increased white adipocyte accumulation in aged male but not female hearts. Adipocyte differentiation is enhanced in isolated stem cell antigen-1 (SCA-1)+ cardiac progenitor cells (CPC) from young male CKO-mice compared with the adipocyte differentiation of male wild-type (WT)-CPC and CPC isolated from female mice. Epigenetic analysis in freshly isolated male CKO-CPC display hypermethylation in pro-angiogenic genes (Fgfr2, Epas1) and hypomethylation in the white adipocyte differentiation gene Zfp423 associated with up-regulated ZFP423 expression and a shift from endothelial to white adipocyte differentiation compared with WT-CPC. The expression of the histone-methyltransferase EZH2 is reduced in male CKO-CPC compared with male WT-CPC, whereas no differences in the EZH2 expression in female CPC were observed. Clonally expanded CPC can differentiate into endothelial cells or into adipocytes depending on the differentiation conditions. ZFP423 overexpression is sufficient to induce white adipocyte differentiation of clonal CPC. In isolated WT-CPC, PGD2 stimulation reduces the expression of EZH2, thereby up-regulating ZFP423 expression and promoting white adipocyte differentiation. The treatment of young male CKO mice with the COX inhibitor Ibuprofen or the PGD2 receptor (DP)2 receptor antagonist BAY-u 3405 in vivo increased EZH2 expression and reduced ZFP423 expression and adipocyte differentiation in CKO-CPC. Thus, cardiomyocyte STAT3 deficiency leads to age-related and sex-specific cardiac remodeling and failure in part due to sex-specific alterations in PGD2 secretion and subsequent epigenetic impairment of the differentiation potential of CPC. Causally involved is the impaired AR signaling in absence of STAT3, which reduces the expression of the PG-degrading enzyme HPGD.


Subject(s)
Myocytes, Cardiac/metabolism , Prostaglandin D2/metabolism , STAT3 Transcription Factor/metabolism , Adipocytes, White/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Cyclooxygenase 2/metabolism , Endothelial Cells/metabolism , Female , Heart Failure/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multipotent Stem Cells/metabolism , Prostaglandin D2/physiology , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Stem Cells/metabolism
6.
Basic Res Cardiol ; 115(6): 62, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32975669

ABSTRACT

Neuraminidase (NEU)1 forms a multienzyme complex with beta-galactosidase (ß-GAL) and protective-protein/cathepsin (PPC) A, which cleaves sialic-acids from cell surface glycoconjugates. We investigated the role of NEU1 in the myocardium after ischemia/reperfusion (I/R). Three days after inducing I/R, left ventricles (LV) of male mice (3 months-old) displayed upregulated neuraminidase activity and increased NEU1, ß-GAL and PPCA expression. Mice hypomorphic for neu1 (hNEU1) had less neuraminidase activity, fewer pro-inflammatory (Lin-CD11b+F4/80+Ly-6Chigh), and more anti-inflammatory macrophages (Lin-CD11b+F4/80+Ly-6Clow) 3 days after I/R, and less LV dysfunction 14 days after I/R. WT mice transplanted with hNEU1-bone marrow (BM) and hNEU1 mice with WT-BM showed significantly better LV function 14 days after I/R compared with WT mice with WT-BM. Mice with a cardiomyocyte-specific NEU1 overexpression displayed no difference in inflammation 3 days after I/R, but showed increased cardiomyocyte hypertrophy, reduced expression and mislocalization of Connexin-43 in gap junctions, and LV dysfunction despite a similar infarct scar size to WT mice 14 days after I/R. The upregulation of NEU1 after I/R contributes to heart failure by promoting inflammation in invading monocytes/macrophages, enhancing cardiomyocyte hypertrophy, and impairing gap junction function, suggesting that systemic NEU1 inhibition may reduce heart failure after I/R.


Subject(s)
Heart Failure/etiology , Hypertrophy, Left Ventricular/etiology , Macrophages/enzymology , Monocytes/enzymology , Myocardial Infarction/complications , Myocardial Reperfusion Injury/complications , Myocytes, Cardiac/enzymology , Neuraminidase/deficiency , Ventricular Dysfunction, Left/etiology , Animals , Cathepsin A/metabolism , Connexin 43/metabolism , Disease Models, Animal , Female , Gap Junctions/enzymology , Gap Junctions/pathology , Heart Failure/enzymology , Heart Failure/immunology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/immunology , Hypertrophy, Left Ventricular/physiopathology , Macrophages/immunology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Myocardial Infarction/enzymology , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Neuraminidase/genetics , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/immunology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Ventricular Remodeling , beta-Galactosidase/metabolism
7.
Leukemia ; 33(6): 1313-1323, 2019 06.
Article in English | MEDLINE | ID: mdl-30546081

ABSTRACT

BCR-ABL+acute lymphoblastic leukemia (ALL) in adults has a poor prognosis with allogeneic stem cell transplantation (SCT) considered the best curative option for suitable patients. We here characterize the curative potential of BH3-mimetics differentially targeting mitochondrial BCL2-family members using a combination therapy approach with dexamethasone and tyrosine kinase inhibitors targeting BCR-ABL. In BCR-ABL + ALL BH3-mimetics act by redistribution of mitochondrial activator BIM, which is strongly required for cytotoxicity of the BCL2-specific BH3-mimetic ABT-199, tyrosine kinase inhibitors (TKIs) and dexamethasone. BIM expression is enhanced by dexamethasone and TKIs and both synergize with ABT-199 in BCR-ABL + ALL. Triple combinations with ABT-199, dexamethasone and TKIs efficiently attenuate leukemia progression both in tissue culture and in primary cell xenotransplantation models. Notably, the dasatinib-containing combination led to treatment- and leukemia-free long-term survival in a BCR-ABL + mouse model. Finally, response to BH3-mimetics can be predicted for individual patients in a clinically relevant setting. These data demonstrate curative targeted and chemotherapy-free pharmacotherapy for BCR-ABL + ALL in a preclinical model. Clinical evaluation, in particular for patients not suitable for allogeneic SCT, is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mitochondria/pathology , Animals , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Dasatinib/administration & dosage , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Mitochondria/metabolism , Sulfonamides/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Eur Heart J ; 38(5): 349-361, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28201733

ABSTRACT

Aims: The benefit of the ß1-adrenergic receptor (ß1-AR) agonist dobutamine for treatment of acute heart failure in peripartum cardiomyopathy (PPCM) is controversial. Cardiac STAT3 expression is reduced in PPCM patients. Mice carrying a cardiomyocyte-restricted deletion of STAT3 (CKO) develop PPCM. We hypothesized that STAT3-dependent signalling networks may influence the response to ß-AR agonist treatment in PPCM patients and analysed this hypothesis in CKO mice. Methods and Results: Follow-up analyses in 27 patients with severe PPCM (left ventricular ejection fraction ≤25%) revealed that 19 of 20 patients not obtaining dobutamine improved cardiac function. All seven patients obtaining dobutamine received heart transplantation (n = 4) or left ventricular assist devices (n = 3). They displayed diminished myocardial triglyceride, pyruvate, and lactate content compared with non-failing controls. The ß-AR agonist isoproterenol (Iso) induced heart failure with high mortality in postpartum female, in non-pregnant female and in male CKO, but not in wild-type mice. Iso induced heart failure and high mortality in CKO mice by impairing fatty acid and glucose uptake, thereby generating a metabolic deficit. The latter was governed by disturbed STAT3-dependent signalling networks, microRNA-199a-5p, microRNA-7a-5p, insulin/glucose transporter-4, and neuregulin/ErbB signalling. The resulting cardiac energy depletion and oxidative stress promoted dysfunction and cardiomyocyte loss inducing irreversible heart failure, which could be attenuated by the ß1-AR blocker metoprolol or glucose-uptake-promoting drugs perhexiline and etomoxir. Conclusions: Iso impairs glucose uptake, induces energy depletion, oxidative stress, dysfunction, and death in STAT3-deficient cardiomyocytes mainly via ß1-AR stimulation. These cellular alterations may underlie the dobutamine-induced irreversible heart failure progression in PPCM patients who frequently display reduced cardiac STAT3 expression.


Subject(s)
Adrenergic beta-1 Receptor Agonists/adverse effects , Adrenergic beta-1 Receptor Agonists/toxicity , Cardiomyopathies/chemically induced , Dobutamine/adverse effects , Heart Failure/drug therapy , Puerperal Disorders/drug therapy , STAT3 Transcription Factor/physiology , Adult , Animals , Blood Glucose/metabolism , Female , Humans , Isoproterenol/pharmacology , Male , Mice, Knockout , MicroRNAs/physiology , Mitochondria, Heart/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Peripartum Period , Purine Nucleotides/metabolism , Random Allocation , Reactive Oxygen Species/metabolism , Receptor, ErbB-4/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/deficiency , Ventricular Dysfunction, Left/chemically induced
9.
Nat Med ; 21(6): 601-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25962121

ABSTRACT

Dysregulation of the actin cytoskeleton in podocytes represents a common pathway in the pathogenesis of proteinuria across a spectrum of chronic kidney diseases (CKD). The GTPase dynamin has been implicated in the maintenance of cellular architecture in podocytes through its direct interaction with actin. Furthermore, the propensity of dynamin to oligomerize into higher-order structures in an actin-dependent manner and to cross-link actin microfilaments into higher-order structures has been correlated with increased actin polymerization and global organization of the actin cytoskeleton in the cell. We found that use of the small molecule Bis-T-23, which promotes actin-dependent dynamin oligomerization and thus increased actin polymerization in injured podocytes, was sufficient to improve renal health in diverse models of both transient kidney disease and CKD. In particular, administration of Bis-T-23 in these renal disease models restored the normal ultrastructure of podocyte foot processes, lowered proteinuria, lowered collagen IV deposits in the mesangial matrix, diminished mesangial matrix expansion and extended lifespan. These results further establish that alterations in the actin cytoskeleton of kidney podocytes is a common hallmark of CKD, while also underscoring the substantial regenerative potential of injured glomeruli and identifying the oligomerization cycle of dynamin as an attractive potential therapeutic target to treat CKD.


Subject(s)
Coumaric Acids/administration & dosage , Cyanoacrylates/administration & dosage , Dynamins/metabolism , Podocytes/drug effects , Proteinuria/drug therapy , Renal Insufficiency, Chronic/drug therapy , Acrylamide/administration & dosage , Actin Cytoskeleton/drug effects , Animals , Dynamins/chemistry , Dynamins/drug effects , Humans , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Mice , Models, Animal , Podocytes/pathology , Podocytes/ultrastructure , Proteinuria/metabolism , Proteinuria/pathology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...