Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 16(2)2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38254822

ABSTRACT

Treatment options for ovarian cancer patients are limited, and a high unmet clinical need remains for targeted and long-lasting, efficient drugs. Genetically modified T cells expressing chimeric antigen receptors (CAR), are promising new drugs that can be directed towards a defined target and have shown efficient, as well as persisting, anti-tumor responses in many patients. We sought to develop novel CAR T cells targeting ovarian cancer and to assess these candidates preclinically. First, we identified potential CAR targets on ovarian cancer samples. We confirmed high and consistent expressions of the tumor-associated antigen FOLR1 on primary ovarian cancer samples. Subsequently, we designed a series of CAR T cell candidates against the identified target and demonstrated their functionality against ovarian cancer cell lines in vitro and in an in vivo xenograft model. Finally, we performed additional in vitro assays recapitulating immune suppressive mechanisms present in solid tumors and developed a process for the automated manufacturing of our CAR T cell candidate. These findings demonstrate the feasibility of anti-FOLR1 CAR T cells for ovarian cancer and potentially other FOLR1-expressing tumors.

2.
Int J Mol Sci ; 24(11)2023 May 24.
Article in English | MEDLINE | ID: mdl-37298141

ABSTRACT

Due to the paucity of targetable antigens, triple-negative breast cancer (TNBC) remains a challenging subtype of breast cancer to treat. In this study, we developed and evaluated a chimeric antigen receptor (CAR) T cell-based treatment modality for TNBC by targeting stage-specific embryonic antigen 4 (SSEA-4), a glycolipid whose overexpression in TNBC has been correlated with metastasis and chemoresistance. To delineate the optimal CAR configuration, a panel of SSEA-4-specific CARs containing alternative extracellular spacer domains was constructed. The different CAR constructs mediated antigen-specific T cell activation characterized by degranulation of T cells, secretion of inflammatory cytokines, and killing of SSEA-4-expressing target cells, but the extent of this activation differed depending on the length of the spacer region. Adoptive transfer of the CAR-engineered T cells into mice with subcutaneous TNBC xenografts mediated a limited antitumor effect but induced severe toxicity symptoms in the cohort receiving the most bioactive CAR variant. We found that progenitor cells in the lung and bone marrow express SSEA-4 and are likely co-targeted by the CAR T cells. Thus, this study has revealed serious adverse effects that raise safety concerns for SSEA-4-directed CAR therapies because of the risk of eliminating vital cells with stem cell properties.


Subject(s)
Receptors, Chimeric Antigen , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Triple Negative Breast Neoplasms/pathology , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , T-Lymphocytes , Xenograft Model Antitumor Assays , Receptors, Antigen, T-Cell , Cell Line, Tumor
3.
Mater Today Bio ; 14: 100259, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35514437

ABSTRACT

Models of heart disease and drug responses are increasingly based on human pluripotent stem cells (hPSCs) since their ability to capture human heart (dys-)function is often better than animal models. Simple monolayer cultures of hPSC-derived cardiomyocytes, however, have shortcomings. Some of these can be overcome using more complex, multi cell-type models in 3D. Here we review modalities that address this, describe efforts to tailor readouts and sensors for monitoring tissue- and cell physiology (exogenously and in situ) and discuss perspectives for implementation in industry and academia.

4.
Sci Rep ; 12(1): 1911, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115587

ABSTRACT

Many critical advances in research utilize techniques that combine high-resolution with high-content characterization at the single cell level. We introduce the MICS (MACSima Imaging Cyclic Staining) technology, which enables the immunofluorescent imaging of hundreds of protein targets across a single specimen at subcellular resolution. MICS is based on cycles of staining, imaging, and erasure, using photobleaching of fluorescent labels of recombinant antibodies (REAfinity Antibodies), or release of antibodies (REAlease Antibodies) or their labels (REAdye_lease Antibodies). Multimarker analysis can identify potential targets for immune therapy against solid tumors. With MICS we analysed human glioblastoma, ovarian and pancreatic carcinoma, and 16 healthy tissues, identifying the pair EPCAM/THY1 as a potential target for chimeric antigen receptor (CAR) T cell therapy for ovarian carcinoma. Using an Adapter CAR T cell approach, we show selective killing of cells only if both markers are expressed. MICS represents a new high-content microscopy methodology widely applicable for personalized medicine.


Subject(s)
Biomarkers, Tumor/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Fluorescent Antibody Technique , Immunotherapy, Adoptive , Neoplasms/metabolism , Neoplasms/therapy , Photobleaching , Single-Cell Analysis , Thy-1 Antigens/metabolism , Cell Death , Cytotoxicity, Immunologic , High-Throughput Screening Assays , Humans , Neoplasms/immunology , Neoplasms/pathology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation
5.
Nat Commun ; 12(1): 1453, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33674603

ABSTRACT

A major roadblock prohibiting effective cellular immunotherapy of pancreatic ductal adenocarcinoma (PDAC) is the lack of suitable tumor-specific antigens. To address this challenge, here we combine flow cytometry screenings, bioinformatic expression analyses and a cyclic immunofluorescence platform. We identify CLA, CD66c, CD318 and TSPAN8 as target candidates among 371 antigens and generate 32 CARs specific for these molecules. CAR T cell activity is evaluated in vitro based on target cell lysis, T cell activation and cytokine release. Promising constructs are evaluated in vivo. CAR T cells specific for CD66c, CD318 and TSPAN8 demonstrate efficacies ranging from stabilized disease to complete tumor eradication with CD318 followed by TSPAN8 being the most promising candidates for clinical translation based on functionality and predicted safety profiles. This study reveals potential target candidates for CAR T cell based immunotherapy of PDAC together with a functional set of CAR constructs specific for these molecules.


Subject(s)
Adenocarcinoma/metabolism , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Immunotherapy/methods , Pancreatic Neoplasms/metabolism , Tetraspanins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Animals , Antigens, Neoplasm/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/therapy , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cytokines/metabolism , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Immunologic Factors , Lymphocyte Activation , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , T-Lymphocytes/immunology , Tetraspanins/genetics , Pancreatic Neoplasms
6.
Artif Organs ; 41(2): 192-199, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27296083

ABSTRACT

Myocardial stem cell therapy in heart failure is strongly dependent on successful cellular transfer, engraftment, and survival. Moreover, massive cell loss directly after intramyocardial injection is commonly observed, generating the need for efficient longitudinal monitoring of transplanted cells in order to develop more efficient transplantation techniques. Therefore, the aim of the present study was to assess viability and cardiac retention of induced pluripotent stem cells after intramyocardial delivery using in vivo bioluminescence analysis (BLI) and magnetic resonance imaging (MRI). Murine induced pluripotent stem cells (iPSCs) were transfected for luciferase reporter gene expression and labeled intracellularly with supraparamagnetic iron oxide particles. Consequently, 5 × 105 cells were transplanted intramyocardially following left anterior descending coronary artery ligation in mice. Cardiac iPSCs were detected using BLI and serial T2* sequences by MRI in a 14-day follow-up. Additionally, infarct extension and left ventricular (LV) function were assessed by MRI. Controls received the same surgical procedure without cell injection. MRI sequences showed a strong MRI signal of labeled iPSCs correlating with myocardial late enhancement, demonstrating engraftment in the infarcted area. Mean iPSC volumes were 4.2 ± 0.4 mm3 at Day 0; 3.1 ± 0.4 mm3 at Day 7; and 5.1 ± 0.8 mm3 after 2 weeks. Thoracic BLI radiance decreased directly after injection from 1.0 × 106 ± 4.2 × 104 (p/s/cm2 /sr) to 1.0 × 105 ± 4.9 × 103 (p/s/cm2 /sr) on Day 1. Afterward, BLI radiance increased to 1.1 × 106 ± 4.2 × 104 (p/s/cm2 /sr) 2 weeks after injection. Cardiac graft localization was confirmed by ex vivo BLI analysis and histology. Left ventricular ejection fraction was higher in the iPSC group (30.9 ± 0.9%) compared to infarct controls (24.0 ± 2.1%; P < 0.05). The combination of MRI and BLI assesses stem cell fate in vivo, enabling cardiac graft localization with evaluation of LV function in myocardial infarction.


Subject(s)
Heart Failure/diagnostic imaging , Heart Failure/therapy , Heart/diagnostic imaging , Induced Pluripotent Stem Cells/transplantation , Animals , Cells, Cultured , Induced Pluripotent Stem Cells/cytology , Luminescent Measurements/methods , Magnetic Resonance Imaging , Mice , Multimodal Imaging/methods , Myocardium/pathology , Optical Imaging/methods
7.
PLoS One ; 10(11): e0143538, 2015.
Article in English | MEDLINE | ID: mdl-26618511

ABSTRACT

RATIONALE: Central questions such as cardiomyocyte subtype emergence during cardiogenesis or the availability of cardiomyocyte subtypes for cell replacement therapy require selective identification and purification of atrial and ventricular cardiomyocytes. However, current methodologies do not allow for a transgene-free selective isolation of atrial or ventricular cardiomyocytes due to the lack of subtype specific cell surface markers. METHODS AND RESULTS: In order to develop cell surface marker-based isolation procedures for cardiomyocyte subtypes, we performed an antibody-based screening on embryonic mouse hearts. Our data indicate that atrial and ventricular cardiomyocytes are characterized by differential expression of integrin α6 (ITGA6) throughout development and in the adult heart. We discovered that the expression level of this surface marker correlates with the intracellular subtype-specific expression of MLC-2a and MLC-2v on the single cell level and thereby enables the discrimination of cardiomyocyte subtypes by flow cytometry. Based on the differential expression of ITGA6 in atria and ventricles during cardiogenesis, we developed purification protocols for atrial and ventricular cardiomyocytes from mouse hearts. Atrial and ventricular identities of sorted cells were confirmed by expression profiling and patch clamp analysis. CONCLUSION: Here, we introduce a non-genetic, antibody-based approach to specifically isolate highly pure and viable atrial and ventricular cardiomyocytes from mouse hearts of various developmental stages. This will facilitate in-depth characterization of the individual cellular subsets and support translational research applications.


Subject(s)
Gene Expression Regulation, Developmental , Heart Atria/cytology , Heart Ventricles/cytology , Integrin alpha6/metabolism , Myocytes, Cardiac/metabolism , Animals , Cells, Cultured , Heart Atria/embryology , Heart Atria/metabolism , Heart Ventricles/embryology , Heart Ventricles/metabolism , Integrin alpha6/genetics , Mice , Myosin Light Chains/genetics , Myosin Light Chains/metabolism
8.
PLoS One ; 9(7): e102171, 2014.
Article in English | MEDLINE | ID: mdl-25029550

ABSTRACT

Cellular reprogramming of somatic cells into induced pluripotent stem cells (iPSC) opens up new avenues for basic research and regenerative medicine. However, the low efficiency of the procedure remains a major limitation. To identify iPSC, many studies to date relied on the activation of pluripotency-associated transcription factors. Such strategies are either retrospective or depend on genetically modified reporter cells. We aimed at identifying naturally occurring surface proteins in a systematic approach, focusing on antibody-targeted markers to enable live-cell identification and selective isolation. We tested 170 antibodies for differential expression between mouse embryonic fibroblasts (MEF) and mouse pluripotent stem cells (PSC). Differentially expressed markers were evaluated for their ability to identify and isolate iPSC in reprogramming cultures. Epithelial cell adhesion molecule (EPCAM) and stage-specific embryonic antigen 1 (SSEA1) were upregulated early during reprogramming and enabled enrichment of OCT4 expressing cells by magnetic cell sorting. Downregulation of somatic marker FAS was equally suitable to enrich OCT4 expressing cells, which has not been described so far. Furthermore, FAS downregulation correlated with viral transgene silencing. Finally, using the marker SSEA-1 we exemplified that magnetic separation enables the establishment of bona fide iPSC and propose strategies to enrich iPSC from a variety of human source tissues.


Subject(s)
Cell Separation/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , fas Receptor/metabolism , Animals , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cell Line , Cellular Reprogramming , Epithelial Cell Adhesion Molecule , Gene Expression Regulation , Humans , Lewis X Antigen/metabolism , Magnetics , Mice
9.
Stem Cell Res ; 10(2): 133-46, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23237958

ABSTRACT

The cellular heterogeneity that is generated during the differentiation of pluripotent stem cells into specific neural subpopulations represents a major obstacle for experimental and clinical progress. To address this problem we developed an optimized strategy for magnetic isolation of PSA-NCAM positive neuronal precursors from embryonic stem cells (ESCs) derived neuronal cultures. PSA-NCAM enrichment at an early step of the in vitro differentiation process increased the number of ES cell derived neurons and reduced cellular diversity. Gene expression analysis revealed that mainly genes involved in neuronal activity were over-represented after purification. In vitro derived PSA-NCAM(+) enriched precursors were characterized in vivo through grafting into the forebrain of adult mice. While unsorted control cells 40 days post graft gave rise to a mixed population composed of immature precursors, early postmitotic neurons and glial cells, PSA-NCAM(+) enriched cells differentiated predominantly into NeuN positive cells. Furthermore, PSA-NCAM enriched population showed efficient migration towards the olfactory bulb after transplantation into the rostral migratory stream of the forebrain neurogenic system. Thus, enrichment of neuronal precursors based on PSA-NCAM expression represents a general and straightforward approach to narrow cellular heterogeneity during neuronal differentiation of pluripotent cells.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Immunomagnetic Separation , Neural Stem Cells/cytology , Neurons/cytology , Actins/metabolism , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Cell Survival/genetics , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/metabolism , Stem Cell Transplantation
10.
Nat Cell Biol ; 13(7): 838-45, 2011 Jun 19.
Article in English | MEDLINE | ID: mdl-21685889

ABSTRACT

Self-renewal of rodent embryonic stem cells is enhanced by partial inhibition of glycogen synthase kinase-3 (Gsk3; refs 1, 2). This effect has variously been attributed to stimulation of Wnt signalling by ß-catenin, stabilization of Myc protein and global de-inhibition of anabolic processes. Here we demonstrate that ß-catenin is not necessary for embryonic stem cell identity or expansion, but its absence eliminates the self-renewal response to Gsk3 inhibition. Responsiveness is fully restored by truncated ß-catenin lacking the carboxy-terminal transactivation domain. However, requirement for Gsk3 inhibition is dictated by expression of T-cell factor 3 (Tcf3) and mediated by direct interaction with ß-catenin. Tcf3 localizes to many pluripotency genes in embryonic stem cells. Our findings confirm that Tcf3 acts as a transcriptional repressor and reveal that ß-catenin directly abrogates Tcf3 function. We conclude that Gsk3 inhibition stabilizes the embryonic stem cell state primarily by reducing repressive influence on the core pluripotency network.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Pluripotent Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Embryonic Stem Cells/enzymology , Gene Expression Regulation, Developmental/drug effects , Gene Regulatory Networks/drug effects , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Mice , Pluripotent Stem Cells/enzymology , RNA Interference , Recombinant Fusion Proteins/metabolism , Transfection , beta Catenin/genetics , beta Catenin/metabolism
11.
Stem Cells ; 27(9): 2069-80, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19544408

ABSTRACT

We have previously demonstrated that differentiation of embryonic stem (ES) cells is associated with downregulation of cell surface E-cadherin. In this study, we assessed the function of E-cadherin in mouse ES cell pluripotency and differentiation. We show that inhibition of E-cadherin-mediated cell-cell contact in ES cells using gene knockout (Ecad(-/-)), RNA interference (EcadRNAi), or a transhomodimerization-inhibiting peptide (CHAVC) results in cellular proliferation and maintenance of an undifferentiated phenotype in fetal bovine serum-supplemented medium in the absence of leukemia inhibitory factor (LIF). Re-expression of E-cadherin in Ecad(-/-), EcadRNAi, and CHAVC-treated ES cells restores cellular dependence to LIF supplementation. Although reversal of the LIF-independent phenotype in Ecad(-/-) ES cells is dependent on the beta-catenin binding domain of E-cadherin, we show that beta-catenin null (betacat(-/-)) ES cells also remain undifferentiated in the absence of LIF. This suggests that LIF-independent self-renewal of Ecad(-/-) ES cells is unlikely to be via beta-catenin signaling. Exposure of Ecad(-/-), EcadRNAi, and CHAVC-treated ES cells to the activin receptor-like kinase inhibitor SB431542 led to differentiation of the cells, which could be prevented by re-expression of E-cadherin. To confirm the role of transforming growth factor beta family signaling in the self-renewal of Ecad(-/-) ES cells, we show that these cells maintain an undifferentiated phenotype when cultured in serum-free medium supplemented with Activin A and Nodal, with fibroblast growth factor 2 required for cellular proliferation. We conclude that transhomodimerization of E-cadherin protein is required for LIF-dependent ES cell self-renewal and that multiple self-renewal signaling networks subsist in ES cells, with activity dependent upon the cellular context.


Subject(s)
Cadherins/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Leukemia Inhibitory Factor/pharmacology , Activins/pharmacology , Animals , Cadherins/genetics , Cadherins/metabolism , Cattle , Cell Communication/drug effects , Cell Communication/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Embryonic Stem Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Mice , Nodal Protein/pharmacology , Protein Multimerization/genetics , Protein Multimerization/physiology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/physiology
12.
Cell Tissue Res ; 327(2): 333-42, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17058052

ABSTRACT

Connexin43 (Cx43) gap-junction channels are highly abundant in intestinal smooth muscle but their functional impact has not been studied so far. Here, we have aimed to elucidate the functional role of Cx43 in the tunica muscularis of the mouse intestine in vivo. Transgenic mice with conditional deletion of Cx43 in smooth muscle cells (SMC) were generated. Histological investigations by immunofluorescence analyses and organ-bath recordings to assess the contractility of intestinal tissue strips were carried out. Measurements of gastrointestinal transit and of the visceromotor response by utilizing a standardized colorectal distension model to quantify alterations of visceral sensory function were also performed in SMC-specific Cx43 null mice and control littermates. Histologically, we found thickening of the tunica muscularis and a 13-fold increase of neutrophil infiltration of the gastrointestinal wall of SMC-specific Cx43 null mice. These animals also exhibited a decrease of 29% in gastrointestinal transit time. In contrast, the visceromotor response to a standardized colorectal distension was elevated, as was the contractility in SMC-specific Cx43 null mice, compared with controls. Thus, SMC-specific ablation of Cx43 in mice leads to morphological and functional alterations of the intestinal tunica muscularis, to gastrointestinal motor dysfunction and to altered visceral sensory function.


Subject(s)
Connexin 43/genetics , Intestinal Mucosa/metabolism , Muscle, Smooth/metabolism , Animals , Body Weight/genetics , Body Weight/physiology , Carbachol/pharmacology , Cell Nucleus/metabolism , Connexin 43/deficiency , Connexin 43/metabolism , Gene Deletion , Gene Expression/drug effects , Inflammation/genetics , Integrases/genetics , Intestines/pathology , Intestines/physiopathology , Lac Operon/genetics , Mice , Mice, Inbred Strains , Mice, Transgenic , Microfilament Proteins/genetics , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle Contraction/physiology , Muscle Proteins/genetics , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Neutrophils/metabolism , Neutrophils/pathology , Pain Measurement , Peroxidase/metabolism , Potassium Chloride/pharmacology , Tamoxifen/pharmacology , beta-Galactosidase/metabolism
13.
J Mol Cell Cardiol ; 41(6): 963-71, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16963078

ABSTRACT

Although the gap junction protein Connexin43 (Cx43) is expressed in various cell types during embryonic development, mice with a global inactivation of Cx43 survive until birth but die perinatally due to an obstruction of the right ventricular outflow tract of the heart. To analyze the functional role of Cx43 gap junction channels in cardiomyocytes of the developing and early postnatal heart, we used alphaMyHC-Cre mice to ablate Cx43 expression selectively in cardiomyocytes during development. We found efficient ablation of Cx43 in cardiomyocytes during embryonic development starting at embryonic day (ED) 9.5 in the ventricular wall. Analyses of cardiac Cx43 protein at birth indicated complete loss of Cx43 expression in cardiomyocytes. All mice homozygously deficient for Cx43 in cardiomyocytes died until postnatal day (PD) 16. Heterozygous inactivation of Cx43 in cardiomyocytes neither altered atrial nor ventricular activation, but homozygous ablation led to changes in ventricular activation, i.e. significant decrease of the QRS-amplitude and prolonged QRS-duration already at PD 4. Cardiac morphology was similar to controls until PD 1, but subtle morphological changes were found in a subgroup of mutant mice at later stages. Besides narrowing of the ventricular outlet region at PD 6, hypertrophy of ventricular myocardium was found at PD 12. Our data indicate that complete inactivation of cardiac Cx43 during development predisposes hearts to develop postnatal morphological alterations, which differ from outflow tract obstructions described for Cx43 null mice. In addition, complete loss of cardiac Cx43 protein during development correlates with slowed ventricular activation at PD 4, impairs viability during development, and leads to death of all mutant mice until PD 16.


Subject(s)
Connexin 43/deficiency , Fetal Heart/embryology , Fetal Heart/metabolism , Myocytes, Cardiac/metabolism , Age Factors , Animals , Connexin 43/genetics , Electrocardiography , Female , Gene Targeting , Gestational Age , Heart/growth & development , Heart/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Pregnancy
14.
J Mol Cell Cardiol ; 41(5): 787-97, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16919680

ABSTRACT

Connexin40 (Cx40) and connexin45 (Cx45) are involved in both cardiac morphogenesis and propagation of electrical activity. We found that Cx40/Cx45 double deficiency (Cx40(-/-)/Cx45(+/-)) causes a variety of cardiac defects leading to high mortality during embryonic development and at birth. The majority of Cx40(-/-)/Cx45(+/-) embryos and postnatal mice suffered from atrioventricular septal defects. Additional cardiac abnormalities, e.g., ventricular septal defects and abnormal myocardial arrangement, occurred at lower abundance. Electrocardiograms of Cx40(-/-)/Cx45(+/+) and Cx40(-/-)/Cx45(+/-) mice revealed prolongation of P-wave, PQ interval and QRS duration compared to controls. Interestingly, in Cx40(-/-)/Cx45(+/-) mice, PQ interval and QRS duration were significantly prolonged compared to Cx40(-/-)/Cx45(+/+) mice. We conclude that the gap junctional proteins Cx40 and Cx45 have overlapping and partially compensatory functions with regard to heart morphogenesis and cardiac conduction. Cx45 might be one of the genetic modifiers that can cause variations in the phenotype of connexin40-deficient animals. Our findings may be particularly relevant for understanding molecular factors contributing to human congenital cardiac diseases.


Subject(s)
Connexins/genetics , Fetal Heart/abnormalities , Heart Conduction System/abnormalities , Heart Defects, Congenital/metabolism , Animals , Connexins/metabolism , Disease Models, Animal , Echocardiography , Fetal Heart/physiopathology , Fetal Viability , Heart Defects, Congenital/chemically induced , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Heterozygote , Homozygote , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphogenesis , Gap Junction alpha-5 Protein
15.
J Cell Sci ; 119(Pt 9): 1715-22, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16595547

ABSTRACT

Gap junctions are characteristically increased in the myometrium during term and preterm delivery and are thought to be essential for the development of uterine contractions during labour. Expression of connexin43 (Cx43), the major myometrial gap junction protein, is increased during delivery. We have generated a mouse mutant (Cx43fl/fl:SM-CreERT2), in which the coding region of Cx43 can be specifically deleted in smooth muscle cells at any given time point by application of tamoxifen. By this approach, we were able to study long-term effects on myometrial functions that are necessary for parturition as well as gap junction intercellular communication in primary myometrial cell cultures. We found a prolongation of the pregnancy in 82% of tamoxifen-treated Cx43fl/fl:SM-CreERT2 mice as well as decreased dye coupling in cultured primary myocytes of these animals. Other parturition-specific parameters such as the regulation of oxytocin receptor, prostaglandin F receptor or progesterone remained unchanged. Our results indicate the important function of Cx43 during parturition in the living animal and suggest further strategies to investigate the role of connexins in uterine contractility in transgenic mice.


Subject(s)
Connexin 43/metabolism , Gap Junctions/metabolism , Myocytes, Smooth Muscle/physiology , Myometrium/cytology , Parturition/physiology , Pregnancy, Animal , Animals , Cell Communication/physiology , Cells, Cultured , Connexin 43/genetics , Estrogen Antagonists/metabolism , Female , Gap Junctions/chemistry , Gene Deletion , Gene Expression Regulation , Genes, fos , Male , Mice , Mice, Knockout , Muscle Contraction/physiology , Myocytes, Smooth Muscle/cytology , Pregnancy , Progesterone/genetics , Progesterone/metabolism , Tamoxifen/metabolism
16.
J Biol Chem ; 280(22): 21594-9, 2005 Jun 03.
Article in English | MEDLINE | ID: mdl-15793309

ABSTRACT

The crucial functions of atrial natriuretic peptide (ANP) and endothelial nitric oxide/NO in the regulation of arterial blood pressure have been emphasized by the hypertensive phenotype of mice with systemic inactivation of either the guanylyl cyclase-A receptor for ANP (GC-A-/-) or endothelial nitric-oxide synthase (eNOS-/-). Intriguingly, similar levels of arterial hypertension are accompanied by marked cardiac hypertrophy in GC-A-/-, but not in eNOS-/-, mice, suggesting that changes in local pathways regulating cardiac growth accelerate cardiac hypertrophy in the former and protect the heart of the latter. Our recent observations in mice with conditional, cardiomyocyte-restricted GC-A deletion demonstrated that ANP locally inhibits cardiomyocyte growth. Abolition of these local, protective effects may enhance the cardiac hypertrophic response of GC-A-/- mice to persistent increases in hemodynamic load. Notably, eNOS-/- mice exhibit markedly increased cardiac ANP levels, suggesting that increased activation of cardiac GC-A can prevent hypertensive heart disease. To test this hypothesis, we generated mice with systemic inactivation of eNOS and cardiomyocyte-restricted deletion of GC-A by crossing eNOS-/- and cardiomyocyte-restricted GC-A-deficient mice. Cardiac deletion of GC-A did not affect arterial hypertension but significantly exacerbated cardiac hypertrophy and fibrosis in eNOS-/- mice. This was accompanied by marked cardiac activation of both the mitogen-activated protein kinase (MAPK) ERK 1/2 and the phosphatase calcineurin. Our observations suggest that local ANP/GC-A/cyclic GMP signaling counter-regulates MAPK/ERK- and calcineurin/nuclear factor of activated T cells-dependent pathways of cardiac myocyte growth in hypertensive eNOS-/- mice.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Myocardium/pathology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/physiology , Animals , Blood Pressure , Blotting, Northern , Blotting, Western , Cyclic GMP/metabolism , Gene Deletion , Genotype , Heart Ventricles/pathology , Hypertension/pathology , Hypertrophy , Mice , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Phenotype , Phosphorylation , RNA, Messenger/metabolism , Signal Transduction
17.
J Cell Sci ; 118(Pt 1): 27-37, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15601660

ABSTRACT

The molecular mechanisms regulating skeletal muscle regeneration and differentiation are not well understood. We analyzed the expression of connexins (Cxs) 40, 43 and 45 in normal and regenerating tibialis anterior muscle and in primary cultures of differentiating myoblasts in adult and newborn mice, respectively. Cxs 45 and 43, but not 40, were strongly expressed in normal muscle and their expression was upregulated during regeneration. Furthermore, the functional role of Cx43 during differentiation and regeneration was examined after induced deletion of Cx43 in transgenic mice. In vivo, the inducible deletion of Cx43 delayed the formation of myofibers and prolonged the expression of myogenin during regeneration. In primary cultures of satellite cell-derived myoblasts, induced deletion of Cx43 led to decreased expression of myogenin and MyoD, dye coupling, creatine kinase activity and myoblast fusion. Thus, the expression of Cx45 and Cx43 is upregulated during skeletal muscle regeneration and Cx43 is required for normal myogenesis in vitro and adult muscle regeneration in vivo.


Subject(s)
Cell Differentiation , Connexin 43/metabolism , Connexins/metabolism , Muscle, Skeletal/physiology , Regeneration , Animals , Animals, Newborn , Blotting, Western , Cell Fusion , Cells, Cultured , Connexin 43/genetics , Connexins/genetics , Creatine Kinase/metabolism , Desmin/metabolism , Female , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation , Male , Mice , Mice, Transgenic , Myoblasts/cytology , Satellite Cells, Skeletal Muscle/cytology , Up-Regulation
18.
Genesis ; 38(4): 159-65, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15083516

ABSTRACT

Conditional gene targeting using the Cre/loxP technology generally includes integration of a selection marker cassette flanked by loxP recognition sites (floxed) in the target gene locus. Subsequent marker removal avoids possible impairment of gene expression or mosaicism due to partial and total deletions after Cre-mediated recombination in vivo. The use of deleter Cre mice for in vivo marker removal in floxed connexin43 mice revealed considerable mosaicism, but no selective marker removal. In addition, we noted that several Cre transgenic lines displayed spontaneous ectopic activity, reminiscent of deleter Cre mice, and required the confirmation of cell type-specific deletion in every individual mouse. When we used myosin heavy chain promoter Cre (alphaMyHC-Cre) mice for cardiomyocyte specific deletion, we observed, in addition to cardiomyocyte-restricted or complete excision, selective marker removal in a subgroup of mice as well. Thus, selective marker removal can be achieved as a byproduct of cell-type restricted deletion.


Subject(s)
Attachment Sites, Microbiological/genetics , Genetic Engineering/methods , Genetic Markers/genetics , Integrases/metabolism , Mutagenesis, Insertional/genetics , Recombination, Genetic/genetics , Sequence Deletion/genetics , Viral Proteins/metabolism , Alleles , Animals , Blotting, Southern , Connexin 26 , Connexin 43/genetics , Connexins/genetics , DNA, Recombinant/genetics , Integrases/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mosaicism , Organ Specificity , Transgenes/genetics , Viral Proteins/genetics
19.
Circulation ; 109(8): 1048-55, 2004 Mar 02.
Article in English | MEDLINE | ID: mdl-14967725

ABSTRACT

BACKGROUND: Connexin 43 (Cx43) is a major determinant of conduction in the ventricular working myocardium of mammals. We investigated the effect of decreased Cx43 expression on conduction velocity and arrhythmogenesis using adult mice with inducible deletion of Cx43. METHODS AND RESULTS: Cx43Cre-ER(T)/+ mice, in which 1 coding region of the Cx43 gene was replaced by Cre-ER(T), were mated to Cx43fl/fl mice, generating Cx43Cre-ER(T)/fl mice. Application of 4-hydroxytamoxifen (4-OHT) induced Cre-ER(T)-mediated deletion of the floxed Cx43 allele. Epicardial ventricular mapping using a 13x19 multiterminal electrode grid (300-microm spacing) was performed on Langendorff-perfused hearts from Cx43fl/fl plus carrier (n=10), Cx43fl/fl plus 4-OHT (n=10), Cx43 Cre-ER(T)/fl plus carrier (n=9), and Cx43Cre-ER(T)/fl plus 4-OHT (n=10). Cx43 protein amount in group 3 hearts was decreased by 50% compared with group 1. 4-OHT did not affect cardiac protein amounts in group 2 but decreased Cx43 expression up to 95% in group 4 compared with group 3. Epicardial activation of both left ventricle (LV) and right ventricle (RV) during sinus rhythm was similar in all groups. Conduction velocity (CV) changed only in group 4 animals. For RV (LV), longitudinal CV decreased from 38 (35) to 31.6 (33.6) and transverse CV from 24.4 (16.8) to 10.1 (11.3) cm/s. Dispersion of conduction in RV (LV) was increased by 91% (38%). Programmed stimulation resulted in ventricular arrhythmias in group 4 (7 of 10 mice) but never in groups 1 through 3. CONCLUSIONS: Heterozygous expression of Cx43 did not affect ventricular conduction velocity. Up to 95% decrease of Cx43 protein in 4-OHT-treated Cx43(Cre-ER(T)/fl) mice reduced conduction velocity and increased dispersion of conduction and propensity for ventricular arrhythmias.


Subject(s)
Connexin 43/deficiency , Heart Conduction System/physiopathology , Tachycardia, Ventricular/etiology , Ventricular Premature Complexes/etiology , Animals , Anisotropy , Collagen/metabolism , Connexin 43/genetics , Connexin 43/physiology , Genotype , Mice , Mice, Knockout , Neural Conduction , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/physiopathology , Ventricular Premature Complexes/genetics , Ventricular Premature Complexes/physiopathology
20.
Cell Commun Adhes ; 10(4-6): 451-6, 2003.
Article in English | MEDLINE | ID: mdl-14681056

ABSTRACT

We analyzed the expression of connexin(Cx)43 in proliferating and differentiating C(2)C(12) cells and in myoblasts obtained from newborn mice. Cx43 was present in both cell types and under both conditions. The functional role of gap junctional communication (GJC) during terminal differentiation was evaluated in C(2)C(12) myoblasts in the presence or absence of the gap junction blocker 18beta-glycyrrhetinic acid (beta-GA). Differentiation was temporally analyzed through myogenin expression, activity of creatine kinase (CK), and yield of multinucleated cells. In cells treated with beta-GA, the CK activity and myotube formation were reversibly blocked. While in control cultures positive myogenin expression was seen in cell clusters, in beta-GA treated cultures the myogenin immunoreactivity was detected in few, preferentially sparse cells. The role of Cx43 during terminal differentiation was evaluated in cultures of myoblasts obtained from Cx43(Cre-ER(T)/fl) transgenic mice. Inducible deletion of Cx43 was obtained upon activation of Cre-ER(T) via 4-OH-tamoxifen applications. Cx43 deletion led to a drastic decrease in myogenin expression at 24 h of differentiation as compared to myoblasts from control mice. Our results indicate that Cx43-containing gap junctions are required for normal skeletal muscle terminal differentiation. These channels might provide a pathway for the intercellular transfer of signals involved in myogenesis.


Subject(s)
Cell Communication/physiology , Cell Differentiation/physiology , Connexin 43/metabolism , Gap Junctions/metabolism , Muscle Development/physiology , Animals , Cell Communication/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Connexin 43/genetics , Creatine Kinase/metabolism , Glycyrrhetinic Acid/pharmacology , Mice , Mice, Transgenic , Muscle Development/drug effects , Muscle Development/genetics , Muscle, Skeletal , Myogenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...