Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Angiogenesis ; 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38965173

RESUMO

Aortic aneurysm is characterized by a pathological dilation at specific predilection sites of the vessel and potentially results in life-threatening vascular rupture. Herein, we established a modified "Häutchen method" for the local isolation of endothelial cells (ECs) from mouse aorta to analyze their spatial heterogeneity and potential role in site-specific disease development. When we compared ECs from aneurysm predilection sites of healthy mice with adjacent control segments we found regulation of genes related to extracellular matrix remodeling, angiogenesis and inflammation, all pathways playing a critical role in aneurysm development. We also detected enhanced cortical stiffness of the endothelium at these sites. Gene expression of ECs from aneurysms of the AngII ApoE-/- model when compared to sham animals mimicked expression patterns from predilection sites of healthy animals. Thus, this work highlights a striking genetic and functional regional heterogeneity in aortic ECs of healthy mice, which defines the location of aortic aneurysm formation in disease.

2.
J Mol Cell Cardiol ; 152: 95-104, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33290769

RESUMO

The adult mammalian heart consists of mononuclear and binuclear cardiomyocytes (CMs) with various ploidies. However, it remains unclear whether a variation in ploidy or number of nuclei is associated with distinct functions and injury responses in CMs, including regeneration. Therefore, we investigated transcriptomes and cellular as well as nuclear features of mononucleated and binucleated CMs in adult mouse hearts with and without injury. To be able to identify the role of ploidy we analyzed control and failing human ventricular CMs because human CMs show a larger and disease-sensitive degree of polyploidization. Using transgenic Myh6-H2BmCh to identify mononucleated and binucleated mouse CMs, we found that cellular volume and RNA content were similar in both. On average nuclei of mononuclear CMs showed a 2-fold higher ploidy, as compared to binuclear CMs indicating that most mononuclear CMs are tetraploid. After myocardial infarction mononucleated and binucleated CMs in the border zone of the lesion responded with hypertrophy and corresponding changes in gene expression, as well as a low level of induction of cell cycle gene expression. Human CMs allowed us to study a wide range of polyploidy spanning from 2n to 16n. Notably, basal as well as pathological gene expression signatures and programs in failing CMs proved to be independent of ploidy. In summary, gene expression profiles were induced in proximity to injury, but independent of number of nuclei or ploidy levels in CMs.


Assuntos
Adaptação Fisiológica , Núcleo Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Ploidias , Regeneração , Animais , Humanos , Masculino , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , RNA-Seq
3.
Basic Res Cardiol ; 110(3): 33, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25925989

RESUMO

Even though the mammalian heart has been investigated for many years, there are still uncertainties in the fields of cardiac cell biology and regeneration with regard to exact fractions of cardiomyocytes (CMs) at different developmental stages, their plasticity after cardiac lesion and also their basal turnover rate. A main shortcoming is the accurate identification of CM and the demonstration of CM division. Therefore, an in vivo model taking advantage of a live reporter-based identification of CM nuclei and their cell cycle status is needed. In this technical report, we describe the generation and characterization of embryonic stem cells and transgenic mice expressing a fusion protein of human histone 2B and the red fluorescence protein mCherry under control of the CM specific αMHC promoter. This fluorescence label allows unequivocal identification and quantitation of CM nuclei and nuclearity in isolated cells and native tissue slices. In ventricles of adults, we determined a fraction of <20 % CMs and binucleation of 77-90 %, while in atria a CM fraction of 30 % and a binucleation index of 14 % were found. We combined this transgenic system with the CAG-eGFP-anillin transgene, which identifies cell division and established a novel screening assay for cell cycle-modifying substances in isolated, postnatal CMs. Our transgenic live reporter-based system enables reliable identification of CM nuclei and determination of CM fractions and nuclearity in heart tissue. In combination with CAG-eGFP-anillin-mice, the cell cycle status of CMs can be monitored in detail enabling screening for proliferation-inducing substances in vitro and in vivo.


Assuntos
Núcleo Celular/metabolismo , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Imagem Óptica/métodos , Animais , Ciclo Celular/fisiologia , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Coração/embriologia , Coração/crescimento & desenvolvimento , Histonas , Humanos , Proteínas Luminescentes , Camundongos , Proteínas Recombinantes de Fusão , Transfecção , Proteína Vermelha Fluorescente
4.
Basic Res Cardiol ; 109(4): 425, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24980781

RESUMO

Ischemic heart disease is associated with inflammation, interstitial fibrosis and ventricular dysfunction prior to the development of heart failure. Endocannabinoids and the cannabinoid receptor CB2 have been claimed to be involved, but their potential role in cardioprotection is not well understood. We therefore explored the role of the cannabinoid receptor CB2 during the initial phase of ischemic cardiomyopathy development prior to the onset of ventricular dysfunction or infarction. Wild type and CB2-deficient mice underwent daily brief, repetitive ischemia and reperfusion (I/R) episodes leading to ischemic cardiomyopathy. The relevance of the endocannabinoid-CB2 receptor axis was underscored by the finding that CB2 was upregulated in ischemic wild type cardiomyocytes and that anandamide level was transiently increased during I/R. CB2-deficient mice showed an increased rate of apoptosis, irreversible loss of cardiomyocytes and persistent left ventricular dysfunction 60 days after the injury, whereas wild type mice presented neither morphological nor functional defects. These defects were due to lack of cardiomyocyte protection mechanisms, as CB2-deficient hearts were in contrast to controls unable to induce switch in myosin heavy chain isoforms, antioxidative enzymes and chemokine CCL2 during repetitive I/R. In addition, a prolonged inflammatory response and adverse myocardial remodeling were found in CB2-deficient hearts because of postponed activation of the M2a macrophage subpopulation. Therefore, the endocannabinoid-CB2 receptor axis plays a key role in cardioprotection during the initial phase of ischemic cardiomyopathy development.


Assuntos
Cardiomiopatias/prevenção & controle , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Transdução de Sinais , Animais , Apoptose , Ácidos Araquidônicos/metabolismo , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Modelos Animais de Doenças , Endocanabinoides/metabolismo , Feminino , Ativação de Macrófagos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/patologia , Alcamidas Poli-Insaturadas/metabolismo , Receptor CB2 de Canabinoide/deficiência , Receptor CB2 de Canabinoide/genética , Fatores de Tempo , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Disfunção Ventricular Esquerda/prevenção & controle , Função Ventricular Esquerda , Remodelação Ventricular
5.
Development ; 137(6): 993-1002, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20179098

RESUMO

beta1 integrins are important regulators of vascular differentiation and development, as their endothelial-specific deletion results in embryonic lethality. In the present study, we investigated the molecular mechanisms underlying the prominent vascular abnormalities that occur in the absence of beta1 integrins. Because of the early embryonic lethality of knockout mice, we studied endothelial cell and vessel development in beta1-integrin-deficient murine embryonic stem cells to gain novel insights into the role of beta1 integrins in vasculo-angiogenesis. We found that vessel development was strongly defective in the mutant embryoid bodies (EBs), as only primitive and short sprouts developed from clusters of vascular precursors in beta1 integrin(-/-) EBs, whereas complex network formation of endothelial tubes was observed in wild-type EBs. The vascular defect was due to deficient beta1 integrin expression in endothelial cells, as its endothelial-specific re-expression rescued the phenotype entirely. The mechanism responsible for defective vessel formation was found to be reduced endothelial cell maturation, migration and elongation. Moreover, the lower number of endothelial cells in beta1 integrin(-/-) EBs was due to an increased apoptosis versus proliferation rate. The enhanced apoptosis and proliferation of beta1 integrin(-/-) endothelial cells was related to the elevation of peNOS and pAKT signaling molecules, respectively. Our data demonstrate that endothelial beta1 integrins are determinants of vessel formation and that this effect is mediated via different signaling pathways.


Assuntos
Vasos Sanguíneos/embriologia , Integrina beta1/fisiologia , Neovascularização Fisiológica/genética , Animais , Apoptose/genética , Vasos Sanguíneos/metabolismo , Movimento Celular/genética , Proliferação de Células , Células Cultivadas , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Endotélio Vascular/embriologia , Endotélio Vascular/fisiologia , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Integrina beta1/genética , Integrina beta1/metabolismo , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo III/fisiologia , Organismos Geneticamente Modificados
6.
J Exp Med ; 203(10): 2315-27, 2006 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-16954371

RESUMO

Cellular cardiomyoplasty is an attractive option for the treatment of severe heart failure. It is, however, still unclear and controversial which is the most promising cell source. Therefore, we investigated and examined the fate and functional impact of bone marrow (BM) cells and embryonic stem cell (ES cell)-derived cardiomyocytes after transplantation into the infarcted mouse heart. This proved particularly challenging for the ES cells, as their enrichment into cardiomyocytes and their long-term engraftment and tumorigenicity are still poorly understood. We generated transgenic ES cells expressing puromycin resistance and enhanced green fluorescent protein cassettes under control of a cardiac-specific promoter. Puromycin selection resulted in a highly purified (>99%) cardiomyocyte population, and the yield of cardiomyocytes increased 6-10-fold because of induction of proliferation on purification. Long-term engraftment (4-5 months) was observed when co-transplanting selected ES cell-derived cardiomyocytes and fibroblasts into the injured heart of syngeneic mice, and no teratoma formation was found (n = 60). Although transplantation of ES cell-derived cardiomyocytes improved heart function, BM cells had no positive effects. Furthermore, no contribution of BM cells to cardiac, endothelial, or smooth muscle neogenesis was detected. Hence, our results demonstrate that ES-based cell therapy is a promising approach for the treatment of impaired myocardial function and provides better results than BM-derived cells.


Assuntos
Células-Tronco Embrionárias/citologia , Contração Miocárdica/fisiologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/transplante , Animais , Transplante de Medula Óssea , Primers do DNA , Eletrofisiologia , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Camundongos , Miócitos Cardíacos/citologia , Puromicina , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Basic Res Cardiol ; 107(2): 257, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22382299

RESUMO

Vessel formation is of critical importance for organ function in the normal and diseased state. In particular, the labeling and quantitation of small vessels prove to be technically challenging using current approaches. We have, therefore, established a transgenic embryonic stem (ES) cell line and a transgenic mouse model where the vascular endothelial growth factor receptor VEGFR-1 (flt-1) promoter drives the expression of the live reporter eGFP. Fluorescence microscopy and immunostainings revealed endothelial-specific eGFP labeling of vascular networks. The expression pattern recapitulates that of the endogenous flt-1 gene, because small and large vessels are labeled by eGFP during embryonic development; after birth, the expression becomes more restricted to small vessels. We have explored this in the cardiovascular system more in detail and found that all small vessels and capillaries within the heart are strongly eGFP+. In addition, myocardial injuries have been induced in transgenic mice and prominent vascular remodeling, and an increase in endothelial cell area within the peri-infarct area could be observed underscoring the utility of this mouse model. Thus, the transgenic flt-1/eGFP models are powerful tools to investigate and quantify vascularization in vivo and to probe the effect of different compounds on vessel formation in vitro.


Assuntos
Endotélio Vascular/citologia , Camundongos Transgênicos , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica/fisiologia , Regiões Promotoras Genéticas , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Western Blotting , Modelos Animais de Doenças , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Camundongos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Cells ; 11(4)2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35203340

RESUMO

Connexins (Cx) are a large family of membrane proteins that can form intercellular connections, so-called gap junctions between adjacent cells. Cx43 is widely expressed in mammals and has a variety of different functions, such as the propagation of electrical conduction in the cardiac ventricle. Despite Cx43 knockout models, many questions regarding the biology of Cx43 in health and disease remain unanswered. Herein we report the establishment of a Cre-inducible Cx43 overexpression system in murine embryonic stem (ES) cells. This enables the investigation of the impact of Cx43 overexpression in somatic cells. We utilized a double reporter system to label Cx43-overexpressing cells via mCherry fluorescence and exogenous Cx43 via fusion with P2A peptide to visualize its distribution pattern. We proved the functionality of our systems in ES cells, HeLa cells, and 3T3-fibroblasts and demonstrated the formation of functional gap junctions based on dye diffusion and FRAP experiments. In addition, Cx43-overexpressing ES cells could be differentiated into viable cardiomyocytes, as shown by the formation of cross striation and spontaneous beating. Analysis revealed faster and more rhythmic beating of Cx43-overexpressing cell clusters. Thus, our Cx43 overexpression systems enable the investigation of Cx43 biology and function in cardiomyocytes and other somatic cells.


Assuntos
Conexina 43 , Células-Tronco Embrionárias Murinas , Animais , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Células HeLa , Humanos , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo
9.
Nat Commun ; 12(1): 3575, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34117258

RESUMO

An amino acid exchange (P209L) in the HSPB8 binding site of the human co-chaperone BAG3 gives rise to severe childhood cardiomyopathy. To phenocopy the disease in mice and gain insight into its mechanisms, we generated humanized transgenic mouse models. Expression of human BAG3P209L-eGFP in mice caused Z-disc disintegration and formation of protein aggregates. This was accompanied by massive fibrosis resulting in early-onset restrictive cardiomyopathy with increased mortality as observed in patients. RNA-Seq and proteomics revealed changes in the protein quality control system and increased autophagy in hearts from hBAG3P209L-eGFP mice. The mutation renders hBAG3P209L less soluble in vivo and induces protein aggregation, but does not abrogate hBAG3 binding properties. In conclusion, we report a mouse model mimicking the human disease. Our data suggest that the disease mechanism is due to accumulation of hBAG3P209L and mouse Bag3, causing sequestering of components of the protein quality control system and autophagy machinery leading to sarcomere disruption.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Cardiomiopatia Restritiva/genética , Cardiomiopatia Restritiva/metabolismo , Animais , Autofagia , Sítios de Ligação , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/terapia , Cardiomiopatia Restritiva/terapia , Criança , Modelos Animais de Doenças , Regulação da Expressão Gênica , Terapia Genética , Coração , Proteínas de Choque Térmico , Humanos , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/metabolismo , Mutação , Ligação Proteica , Proteômica , Sarcômeros/metabolismo
10.
Cell Death Differ ; 27(4): 1398-1414, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31570857

RESUMO

Despite the great progress on the cell biology of programmed cell death (PCD), its incidence and exact time course during embryonic and particular heart development are still unclear. This is also due to the lack of models enabling to directly identify and monitor PCD cells at different time points in vivo. Herein we report generation of transgenic murine embryonic stem cell and mouse models expressing secreted Annexin V-YFP under control of the CAG promoter. This enables to visualize and quantify PCD in vitro and in vivo during embryonic development. At early embryonic stages we found Annexin V-YFP+ fluorescent cells in known areas of PCD, such as the otic ring and at the site of neural tube closing, underscoring its specificity for detection of PCD. We have focused our detailed analysis primarily on PCD in the embryonic heart for a better understanding of its role during development. Our findings reveal that PCD peaks at early stages of cardiogenesis (E9.5-E13.5) and strongly decreases thereafter. Moreover, the PCD cells in the heart are predominantly cardiomyocytes, and an unexpected area of prominent cardiac PCD are the ventricular trabeculae (E9.5-E14.5). Thus, the sA5-YFP mouse line provides novel insight into the incidence and relevance of cardiac PCD during embryonic development ex- and in vivo.


Assuntos
Apoptose , Coração/embriologia , Animais , Caspases/metabolismo , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Genes Reporter , Camundongos , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/citologia , Tubo Neural/embriologia , Organogênese
11.
Cell Stem Cell ; 27(1): 50-63.e5, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32619518

RESUMO

Modulating signaling pathways including Wnt and Hippo can induce cardiomyocyte proliferation in vivo. Applying these signaling modulators to human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in vitro can expand CMs modestly (<5-fold). Here, we demonstrate massive expansion of hiPSC-CMs in vitro (i.e., 100- to 250-fold) by glycogen synthase kinase-3ß (GSK-3ß) inhibition using CHIR99021 and concurrent removal of cell-cell contact. We show that GSK-3ß inhibition suppresses CM maturation, while contact removal prevents CMs from cell cycle exit. Remarkably, contact removal enabled 10 to 25 times greater expansion beyond GSK-3ß inhibition alone. Mechanistically, persistent CM proliferation required both LEF/TCF activity and AKT phosphorylation but was independent from yes-associated protein (YAP) signaling. Engineered heart tissues from expanded hiPSC-CMs showed comparable contractility to those from unexpanded hiPSC-CMs, demonstrating uncompromised cellular functionality after expansion. In summary, we uncovered a molecular interplay that enables massive hiPSC-CM expansion for large-scale drug screening and tissue engineering applications.


Assuntos
Células-Tronco Pluripotentes Induzidas , Diferenciação Celular , Células Cultivadas , Glicogênio Sintase Quinase 3 beta , Humanos , Miócitos Cardíacos
12.
Cell Stem Cell ; 22(2): 262-276.e7, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29451855

RESUMO

Despite much work studying ex vivo multipotent stromal cells (MSCs), the identity and characteristics of MSCs in vivo are not well defined. Here, we generated a CD73-EGFP reporter mouse to address these questions and found EGFP+ MSCs in various organs. In vivo, EGFP+ mesenchymal cells were observed in fetal and adult bones at proliferative ossification sites, while in solid organs EGFP+ cells exhibited a perivascular distribution pattern. EGFP+ cells from the bone compartment could be clonally expanded ex vivo from single cells and displayed trilineage differentiation potential. Moreover, in the central bone marrow CD73-EGFP+ specifically labeled sinusoidal endothelial cells, thought to be a critical component of the hematopoietic stem cell niche. Purification and molecular characterization of this CD73-EGFP+ population revealed an endothelial subtype that also displays a mesenchymal signature, highlighting endothelial cell heterogeneity in the marrow. Thus, the CD73-EGFP mouse is a powerful tool for studying MSCs and sinusoidal endothelium.


Assuntos
5'-Nucleotidase/metabolismo , Células da Medula Óssea/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Multipotentes/metabolismo , Coloração e Rotulagem , Nicho de Células-Tronco , Animais , Medula Óssea/metabolismo , Células da Medula Óssea/citologia , Condrogênese , Células Endoteliais/citologia , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Especificidade de Órgãos , Células Estromais/citologia , Células Estromais/metabolismo
13.
Sci Rep ; 8(1): 7145, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29739982

RESUMO

Ventricular tachycardia (VT) is the most common and potentially lethal complication following myocardial infarction (MI). Biological correction of the conduction inhomogeneity that underlies re-entry could be a major advance in infarction therapy. As minimal increases in conduction of infarcted tissue markedly influence VT susceptibility, we reasoned that enhanced propagation of the electrical signal between non-excitable cells within a resolving infarct might comprise a simple means to decrease post-infarction arrhythmia risk. We therefore tested lentivirus-mediated delivery of the gap-junction protein Connexin 43 (Cx43) into acute myocardial lesions. Cx43 was expressed in (myo)fibroblasts and CD45+ cells within the scar and provided prominent and long lasting arrhythmia protection in vivo. Optical mapping of Cx43 injected hearts revealed enhanced conduction velocity within the scar, indicating Cx43-mediated electrical coupling between myocytes and (myo)fibroblasts. Thus, Cx43 gene therapy, by direct in vivo transduction of non-cardiomyocytes, comprises a simple and clinically applicable biological therapy that markedly reduces post-infarction VT.


Assuntos
Arritmias Cardíacas/genética , Cicatriz/genética , Conexina 43/genética , Terapia Genética , Infarto do Miocárdio/genética , Animais , Arritmias Cardíacas/complicações , Arritmias Cardíacas/patologia , Arritmias Cardíacas/terapia , Cicatriz/patologia , Cicatriz/terapia , Conexina 43/administração & dosagem , Modelos Animais de Doenças , Fibroblastos/metabolismo , Vetores Genéticos/uso terapêutico , Células HEK293 , Humanos , Lentivirus/genética , Camundongos , Células Musculares/metabolismo , Células Musculares/patologia , Mioblastos/metabolismo , Mioblastos/patologia , Infarto do Miocárdio/complicações , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Taquicardia Ventricular/complicações , Taquicardia Ventricular/genética , Taquicardia Ventricular/patologia , Taquicardia Ventricular/terapia
14.
Clin Cancer Res ; 11(16): 5993-6001, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16115944

RESUMO

PURPOSE: We conducted a phase I/II randomized trial to evaluate the clinical and immunologic effect of chemotherapy combined with vaccination in primary metastatic colorectal cancer patients with a carcinoembryonic antigen-derived peptide in the setting of adjuvants granulocyte macrophage colony-stimulating factor, CpG-containing DNA molecules (dSLIM), and dendritic cells. EXPERIMENTAL DESIGN: HLA-A2-positive patients with confirmed newly diagnosed metastatic colorectal cancer and elevated serum carcinoembryonic antigen (CEA) were randomized to receive three cycles of standard chemotherapy (irinotecan/high-dose 5-fluorouracil/leucovorin) and vaccinations with CEA-derived CAP-1 peptide admixed with different adjuvants [CAP-1/granulocyte macrophage colony-stimulating factor/interleukin-2 (IL-2), CAP-1/dSLIM/IL-2, and CAP-1/IL-2]. After completion of chemotherapy, patients received weekly vaccinations until progression of disease. Immune assessment was done at baseline and after three cycles of combined chemoimmunotherapy. HLA-A2 tetramers complexed with the peptides CAP-1, human T-cell lymphotrophic virus type I TAX, cytomegalovirus (CMV) pp65, and EBV BMLF-1 were used for phenotypic immune assessment. IFN-gamma intracellular cytokine assays were done to evaluate CTL reactivity. RESULTS: Seventeen metastatic patients were recruited, of whom 12 completed three cycles. Therapy resulted in five complete response, one partial response, five stable disease, and six progressive disease. Six grade 1 local skin reactions and one mild systemic reaction to vaccination treatment were observed. Overall survival after a median observation time of 29 months was 17 months with a survival rate of 35% (6 of 17) at that time. Eight patients (47%) showed elevation of CAP-1-specific CTLs. Neither of the adjuvants provided superiority in eliciting CAP-1-specific immune responses. During three cycles of chemotherapy, EBV/CMV recall antigen-specific CD8+ cells decreased by an average 14%. CONCLUSIONS: The presented chemoimmunotherapy is a feasible and safe combination therapy with clinical and immunologic efficacy. Despite concurrent chemotherapy, increases in CAP-1-specific T cells were observed in 47% of patients after vaccination.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígeno Carcinoembrionário/imunologia , Neoplasias Colorretais/terapia , Antígeno HLA-A2/imunologia , Oligopeptídeos/uso terapêutico , Adulto , Idoso , Complexo CD3/análise , Antígenos CD4/análise , Antígenos CD8/análise , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Antígeno Carcinoembrionário/sangue , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Terapia Combinada , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Fluoruracila/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Humanos , Epitopos Imunodominantes/imunologia , Imunoterapia/métodos , Interferon gama/metabolismo , Interleucina-2/administração & dosagem , Irinotecano , Leucovorina/administração & dosagem , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Masculino , Pessoa de Meia-Idade , Oligopeptídeos/administração & dosagem , Oligopeptídeos/imunologia , Estudos Prospectivos , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Resultado do Tratamento
15.
PLoS One ; 8(7): e70543, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936225

RESUMO

Embryonic stem cells (ES) and induced pluripotent stem (iPS) cells represent promising tools for cell-based therapies and regenerative medicine. Nevertheless, implantation of ES cell derived differentiated cells holds the risk of teratoma formation due to residual undifferentiated cells. In order to tackle this problem, we used pluripotent stem cells consisting of ES and iPS cells of mouse genetically modified by lentiviral vectors (LVs) carrying herpes simplex virus thymidine kinase (HSV-TK) under the control of different promoters of pluripotency genes. Cells expressing TK in turn are eliminated upon administration of the prodrug ganciclovir (GCV). Our aim was to study the conditions required for a safe mechanism to clear residual undifferentiated cells but using low MOIs of lentiviruses to reduce the risk of insertional mutagenesis. Our in vitro data demonstrated that TK expression in pluripotent stem cells upon treatment with GCV led to elimination of undifferentiated cells. However, introduction of hygromycin resistance in the LV transduced ES cells followed by pre-selection with hygromycin and GCV treatment was required to abolish undifferentiated cells. Most importantly, transplantation of pre-selected ES cells that had been transduced with low MOI LV in mice resulted in no teratoma development after GCV treatment in vivo. Taken together, our data show that pre-selection of ES cells prior to in vivo application is necessary if vector integration events are minimized. The study presented here gives rise to safer use of pluripotent stem cells as promising cell sources in regenerative medicine in the future.


Assuntos
Regulação da Expressão Gênica , Vetores Genéticos/genética , Lentivirus/genética , Células-Tronco Pluripotentes/metabolismo , Timidina Quinase/genética , Timidina Quinase/metabolismo , Animais , Efeito Espectador , Linhagem Celular , Modelos Animais de Doenças , Células-Tronco Embrionárias/metabolismo , Ganciclovir/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Ordem dos Genes , Genes Transgênicos Suicidas , Terapia Genética , Camundongos , Células-Tronco Pluripotentes/transplante , Pró-Fármacos/farmacologia , Regiões Promotoras Genéticas , Simplexvirus/enzimologia , Transplante de Células-Tronco , Teratoma/genética , Teratoma/patologia , Teratoma/terapia , Transdução Genética
16.
Nat Cell Biol ; 10(5): 584-92, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18425115

RESUMO

Recent studies have suggested that regeneration of non-haematopoietic cell lineages can occur through heterotypic cell fusion with haematopoietic cells of the myeloid lineage. Here we show that lymphocytes also form heterotypic-fusion hybrids with cardiomyocytes, skeletal muscle, hepatocytes and Purkinje neurons. However, through lineage fate-mapping we demonstrate that such in vivo fusion of lymphoid and myeloid blood cells does not occur to an appreciable extent in steady-state adult tissues or during normal development. Rather, fusion of blood cells with different non-haematopoietic cell types is induced by organ-specific injuries or whole-body irradiation, which has been used in previous studies to condition recipients of bone marrow transplants. Our findings demonstrate that blood cells of the lymphoid and myeloid lineages contribute to various non-haematopoietic tissues by forming rare fusion hybrids, but almost exclusively in response to injuries or inflammation.


Assuntos
Fusão Celular , Linhagem da Célula , Células-Tronco Hematopoéticas/fisiologia , Linfócitos/fisiologia , Células Mieloides/fisiologia , Radiação , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Hematopoéticas/citologia , Hepatócitos/citologia , Hepatócitos/fisiologia , Linfócitos/citologia , Camundongos , Camundongos SCID , Camundongos Transgênicos , Músculo Esquelético/citologia , Células Mieloides/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Células de Purkinje/citologia , Células de Purkinje/fisiologia , Quimeras de Transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA