Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Cell Mol Life Sci ; 80(9): 265, 2023 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-37615749

RESUMO

Transient receptor potential cation channel-6 (TRPC6) gene mutations cause familial focal segmental glomerulosclerosis (FSGS), which is inherited as an autosomal dominant disease. In patients with TRPC6-related FSGS, all mutations map to the N- or C-terminal TRPC6 protein domains. Thus far, the majority of TRPC6 mutations are missense resulting in increased or decreased calcium influx; however, the fundamental molecular mechanisms causing cell injury and kidney pathology are unclear. We report a novel heterozygous TRPC6 mutation (V691Kfs*) in a large kindred with no signs of FSGS despite a largely truncated TRPC6 protein. We studied the molecular effects of V691Kfs* TRPC6 mutant using the tridimensional cryo-EM structure of the tetrameric TRPC6 protein. The results indicated that V691 is localized at the pore-forming transmembrane region affecting the ion conduction pathway, and predicted that V691Kfs* causes closure of the ion-conducting pathway leading to channel inactivation. We assessed the impact of V691Kfs* and two previously reported TRPC6 disease mutants (P112Q and G757D) on calcium influx in cells. Our data show that the V691Kfs* fully inactivated the TRCP6 channel-specific calcium influx consistent with a complete loss-of-function phenotype. Furthermore, the V691Kfs* truncation exerted a dominant negative effect on the full-length TRPC6 proteins. In conclusion, the V691Kfs* non-functional truncated TRPC6 is not sufficient to cause FSGS. Our data corroborate recently characterized TRPC6 loss-of-function and gain-of-function mutants suggesting that one defective TRPC6 gene copy is not sufficient to cause FSGS. We underscore the importance of increased rather than reduced calcium influx through TRPC6 for podocyte cell death.


Assuntos
Glomerulosclerose Segmentar e Focal , Humanos , Glomerulosclerose Segmentar e Focal/genética , Canal de Cátion TRPC6/genética , Cálcio , Mutação com Perda de Função , Mutação/genética
2.
Int J Mol Sci ; 22(2)2021 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-33478148

RESUMO

Although cardiovascular devices are mostly implanted in arteries or to replace arteries, in vitro studies on implant endothelialization are commonly performed with human umbilical cord-derived venous endothelial cells (HUVEC). In light of considerable differences, both morphologically and functionally, between arterial and venous endothelial cells, we here compare HUVEC and human umbilical cord-derived arterial endothelial cells (HUAEC) regarding their equivalence as an endothelial cell in vitro model for cardiovascular research. No differences were found in either for the tested parameters. The metabolic activity and lactate dehydrogenase, an indicator for the membrane integrity, slightly decreased over seven days of cultivation upon normalization to the cell number. The amount of secreted nitrite and nitrate, as well as prostacyclin per cell, also decreased slightly over time. Thromboxane B2 was secreted in constant amounts per cell at all time points. The Von Willebrand factor remained mainly intracellularly up to seven days of cultivation. In contrast, collagen and laminin were secreted into the extracellular space with increasing cell density. Based on these results one might argue that both cell types are equally suited for cardiovascular research. However, future studies should investigate further cell functionalities, and whether arterial endothelial cells from implantation-relevant areas, such as coronary arteries in the heart, are superior to umbilical cord-derived endothelial cells.


Assuntos
Pesquisa Biomédica , Doenças Cardiovasculares/terapia , Células Endoteliais da Veia Umbilical Humana/citologia , Artérias Umbilicais/citologia , Implantes Absorvíveis , Citoesqueleto de Actina/metabolismo , Pesquisa Biomédica/métodos , Pesquisa Biomédica/tendências , Doenças Cardiovasculares/etiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Baseada em Transplante de Células e Tecidos/tendências , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Medicina Regenerativa/métodos , Medicina Regenerativa/tendências , Engenharia Tecidual/métodos , Engenharia Tecidual/tendências , Artérias Umbilicais/metabolismo , Fator de von Willebrand/metabolismo
3.
Int J Mol Sci ; 22(23)2021 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-34884923

RESUMO

Insufficient endothelialization of cardiovascular grafts is a major hurdle in vascular surgery and regenerative medicine, bearing a risk for early graft thrombosis. Neither of the numerous strategies pursued to solve these problems were conclusive. Endothelialization is regulated by the endothelial basement membrane (EBM), a highly specialized part of the vascular extracellular matrix. Thus, a detailed understanding of the structure-function interrelations of the EBM components is fundamental for designing biomimetic materials aiming to mimic EBM functions. In this review, a detailed description of the structure and functions of the EBM are provided, including the luminal and abluminal interactions with adjacent cell types, such as vascular smooth muscle cells. Moreover, in vivo as well as in vitro strategies to build or renew EBM are summarized and critically discussed. The spectrum of methods includes vessel decellularization and implant biofunctionalization strategies as well as tissue engineering-based approaches and bioprinting. Finally, the limitations of these methods are highlighted, and future directions are suggested to help improve future design strategies for EBM-inspired materials in the cardiovascular field.


Assuntos
Membrana Basal/química , Membrana Basal/metabolismo , Prótese Vascular , Endotélio Vascular/citologia , Animais , Materiais Biocompatíveis , Bioimpressão , Matriz Extracelular , Humanos , Miócitos de Músculo Liso , Desenho de Prótese , Engenharia Tecidual/métodos
4.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34209789

RESUMO

Near-physiological in vitro thrombogenicity test systems for the evaluation of blood-contacting endothelialized biomaterials requires co-cultivation with platelets (PLT). However, the addition of PLT has led to unphysiological endothelial cell (EC) detachment in such in vitro systems. A possible cause for this phenomenon may be PLT activation triggered by the applied endothelial cell medium, which typically consists of basal medium (BM) and nine different supplements. To verify this hypothesis, the influence of BM and its supplements was systematically analyzed regarding PLT responses. For this, human platelet rich plasma (PRP) was mixed with BM, BM containing one of nine supplements, or with BM containing all supplements together. PLT adherence analysis was carried out in six-channel slides with plasma-treated cyclic olefin copolymer (COC) and poly(tetrafluoro ethylene) (PTFE, as a positive control) substrates as part of the six-channel slides in the absence of EC and under static conditions. PLT activation and aggregation were analyzed using light transmission aggregometry and flow cytometry (CD62P). Medium supplements had no effect on PLT activation and aggregation. In contrast, supplements differentially affected PLT adherence, however, in a polymer- and donor-dependent manner. Thus, the use of standard endothelial growth medium (BM + all supplements) maintains functionality of PLT under EC compatible conditions without masking the differences of PLT adherence on different polymeric substrates. These findings are important prerequisites for the establishment of a near-physiological in vitro thrombogenicity test system assessing polymer-based cardiovascular implant materials in contact with EC and PLT.


Assuntos
Materiais Biocompatíveis/farmacologia , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Meios de Cultura/farmacologia , Adulto , Materiais Biocompatíveis/química , Plaquetas/citologia , Meios de Cultura/química , Endotélio/citologia , Feminino , Humanos , Masculino , Teste de Materiais , Pessoa de Meia-Idade , Ativação Plaquetária/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Polímeros/farmacologia , Alicerces Teciduais/química
5.
J Transl Med ; 18(1): 437, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33208161

RESUMO

BACKGROUND: Vectors derived from adeno-associated viruses (AAVs) are widely used for gene transfer both in vitro and in vivo and have gained increasing interest as shuttle systems to deliver therapeutic genes to the heart. However, there is little information on their tissue penetration and cytotoxicity, as well as the optimal AAV serotype for transferring genes to diseased hearts. Therefore, we aimed to establish an organotypic heart slice culture system for mouse left ventricular (LV) myocardium and use this platform to analyze gene transfer efficiency, cell tropism, and toxicity of different AAV serotypes. METHODS: LV tissue slices, 300 µm thick, were prepared from 15- to 17-day-old transgenic alpha-myosin heavy-chain-mCherry mice using a vibrating microtome. Tissue slice viability in air-liquid culture was evaluated by calcein-acetoxymethyl ester staining, mCherry fluorescence intensity, and the tetrazolium assay. Four recombinant AAV serotypes (1, 2, 6, 8) expressing green fluorescent protein (GFP) under the CAG promoter were added to the slice surface. Gene transfer efficiency was quantified as the number of GFP-positive cells per slice. AAV cell tropism was examined by comparing the number of GFP-positive cardiomyocytes (CMs) and fibroblasts within heart slices. RESULTS: Slices retained viability in in vitro culture for at least 5 days. After adding AAV particles, AAV6-infected slices showed the highest number of GFP-expressing cells, almost exclusively CMs. Slice incubation with AAV1, 2, and 8 resulted in fewer GFP-positive cells, with AAV2 having the lowest gene transfer efficiency. None of the AAV serotypes tested caused significant cytotoxicity when compared to non-infected control slices. CONCLUSIONS: We have established a readily available mouse organotypic heart slice culture model and provided evidence that AAV6 may be a promising gene therapy vector for heart failure and other cardiac diseases.


Assuntos
Dependovirus , Terapia Genética , Animais , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Sorogrupo , Transdução Genética
6.
FASEB J ; 33(1): 49-70, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30188756

RESUMO

To date, no viable therapeutic options exist for the effective and sustained reversal of cardiac failure, other than heart transplantation and mechanical circulatory assist devices. Therefore, divergent strategies aiming at the de novo formation of contractile tissue, as a prerequisite for the restoration of cardiac pump function, are currently being pursued. Clinical trials involving the transplantation of somatic progenitor cells failed. The search for alternative cell-based strategies to combat the consequences of ischemic injury has sparked widespread interest in the genetic and pharmacologic reprogramming of fibroblasts into cardiomyocytes, harnessing the abundant in vivo pool of cardiac fibroblasts. Here, we provide a comprehensive overview of in vitro and in vivo cardiac reprogramming studies identified in an extensive literature search. We systematically review and evaluate feasibility, efficiency, and reproducibility of the different technologies currently being explored. Finally, we discuss potential safety issues deduced from preclinical studies and identify obstacles that must be overcome before clinical translation.-Klose, K., Gossen, M., Stamm, C. Turning fibroblasts into cardiomyocytes: technological review of cardiac transdifferentiation strategies.


Assuntos
Transdiferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Reprogramação Celular , Fibroblastos/citologia , Insuficiência Cardíaca/terapia , Miócitos Cardíacos/citologia , Regeneração , Animais , Humanos
7.
Thorac Cardiovasc Surg ; 66(1): 53-62, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29216651

RESUMO

For more than 20 years, tremendous efforts have been made to develop cell-based therapies for treatment of heart failure. However, the results of clinical trials using somatic, nonpluripotent stem or progenitor cells have been largely disappointing in both cardiology and cardiac surgery scenarios. Surgical groups were among the pioneers of experimental and clinical myocyte transplantation ("cellular cardiomyoplasty"), but little translational progress was made prior to the development of cellular reprogramming for creation of induced pluripotent stem cells (iPSC). Ever since, protocols have been developed which allow for the derivation of large numbers of autologous cardiomyocytes (CMs) from patient-specific iPSC, moving translational research closer toward clinical pilot trials. However, compared with somatic cell therapy, the technology required for safe and efficacious pluripotent stem cell (PSC)-based therapies is extremely complex and requires tremendous resources and close interactions between basic scientists and clinicians. This review summarizes PSC sources, strategies to derive CMs, current cardiac tissue engineering approaches, concerns regarding immunogenicity and cellular maturity, and highlights the contributions made by surgical groups.


Assuntos
Doenças Cardiovasculares/cirurgia , Células-Tronco Embrionárias/transplante , Miocárdio/patologia , Miócitos Cardíacos/transplante , Células-Tronco Pluripotentes/transplante , Regeneração , Medicina Regenerativa/métodos , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Linhagem da Célula , Reprogramação Celular , Técnicas de Reprogramação Celular , Células-Tronco Embrionárias/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Recuperação de Função Fisiológica , Transdução de Sinais , Resultado do Tratamento
8.
Biomacromolecules ; 18(11): 3819-3833, 2017 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-28954190

RESUMO

The rational design of a polyplex gene carrier aims to balance maximal effectiveness of nucleic acid transfection into cells with minimal adverse effects. Depsipeptide blocks with an Mn ∼ 5 kDa exhibiting strong physical interactions were conjugated with PEI moieties (2.5 or 10 kDa) to di- and triblock copolymers. Upon nanoparticle formation and complexation with DNA, the resulting polyplexes (sizes typically 60-150 nm) showed remarkable stability compared to PEI-only or lipoplex and facilitated efficient gene delivery. Intracellular trafficking was visualized by observing fluorescence-labeled pDNA and highlighted the effective cytoplasmic uptake of polyplexes and release of DNA to the perinuclear space. Specifically, a triblock copolymer with a middle depsipeptide block and two 10 kDa PEI swallowtail structures mediated the highest levels of transgenic VEGF secretion in mesenchymal stem cells with low cytotoxicity. These nanocarriers form the basis for a delivery platform technology, especially for gene transfer to primary human cells.


Assuntos
DNA/genética , Depsipeptídeos/química , Técnicas de Transferência de Genes , Nanopartículas/química , Sobrevivência Celular/genética , DNA/química , Depsipeptídeos/genética , Humanos , Plasmídeos/química , Plasmídeos/genética , Polietilenoglicóis/química , Polietilenoimina/química , Cultura Primária de Células , Transfecção/métodos
9.
Cell Mol Life Sci ; 72(23): 4671-80, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26109426

RESUMO

Human induced pluripotent stem cells (hiPSC) differentiate into multiple cell types. Selective cell targeting is often needed for analyzing gene function by overexpressing proteins in a distinct population of hiPSC-derived cell types and for monitoring cell fate in response to stimuli. However, to date, this has not been possible, as commonly used viruses enter the hiPSC via ubiquitously expressed receptors. Here, we report for the first time the application of a heterologous avian receptor, the tumor virus receptor A (TVA), to selectively transduce TVA(+) cells in a mixed cell population. Expression of the TVA surface receptor via genetic engineering renders cells susceptible for infection by avian leucosis virus (ALV). We generated hiPSC lines with this stably integrated, ectopic TVA receptor gene that expressed the receptor while retaining pluripotency. The undifferentiated hiPSC(TVA+) as well as their differentiating progeny could be infected by recombinant ALV (so-called RCAS virus) with high efficiency. Due to incomplete receptor blocking, even sequential infection of differentiating or undifferentiated TVA(+) cells was possible. In conclusion, the TVA/RCAS system provides an efficient and gentle gene transfer system for hiPSC and extends our possibilities for selective cell targeting and lineage tracing studies.


Assuntos
Proteínas Aviárias/genética , Vírus do Sarcoma Aviário/genética , Engenharia Genética/métodos , Células-Tronco Pluripotentes Induzidas/virologia , Receptores Virais/genética , Proteínas Aviárias/metabolismo , Vírus do Sarcoma Aviário/patogenicidade , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Elementos de DNA Transponíveis , Citometria de Fluxo/métodos , Vetores Genéticos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Receptores Virais/metabolismo
10.
Nucleic Acids Res ; 42(21): 13061-73, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25389273

RESUMO

Engineered transcription activator-like effectors, or TALEs, have emerged as a new class of designer DNA-binding proteins. Their DNA recognition sites can be specified with great flexibility. When fused to appropriate transcriptional regulatory domains, they can serve as designer transcription factors, modulating the activity of targeted promoters. We created tet operator (tetO)-specific TALEs (tetTALEs), with an identical DNA-binding site as the Tet repressor (TetR) and the TetR-based transcription factors that are extensively used in eukaryotic transcriptional control systems. Different constellations of tetTALEs and tetO modified chromosomal transcription units were analyzed for their efficacy in mammalian cells. We find that tetTALE-silencers can entirely abrogate expression from the strong human EF1α promoter when binding upstream of the transcriptional control sequence. Remarkably, the DNA-binding domain of tetTALE alone can effectively counteract trans-activation mediated by the potent tettrans-activator and also directly interfere with RNA polymerase II transcription initiation from the strong CMV promoter. Our results demonstrate that TALEs can act as highly versatile tools in genetic engineering, serving as trans-activators, trans-silencers and also competitive repressors.


Assuntos
Regulação da Expressão Gênica , Proteínas Repressoras/metabolismo , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Citomegalovirus/genética , Humanos , Regiões Promotoras Genéticas , Engenharia de Proteínas , Proteínas Repressoras/química , Transativadores/metabolismo
11.
Cytotherapy ; 17(2): 199-214, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25457280

RESUMO

BACKGROUND AIMS: In Parkinson's disease (PD), neurogenesis in the subventricular zone (SVZ)-olfactory bulb (OB) axis is affected as the result of the lack of dopaminergic innervations reaching the SVZ. This aberrant network has been related to the hyposmia of PD patients, which is an early diagnostic marker of the disease. Consequently, much interest arose in finding mechanisms to modulate the SVZ-OB axis. Direct modulation of this axis could be achieved by transplantation of mesenchymal stromal cells (MSC), as it has been shown in rat and mouse PD models. However, the neurogenic effect of MSC in PD was thus far only analyzed weeks after transplantation, and little is known about effects immediately after transplantation. METHODS: We assessed the acute neuroprotective and neurogenic effects of adipose-derived MSC transplanted into the rat substantia nigra in the 6-hydroxydopamine model of PD. RESULTS: Three days after transplantation, subventricular neurogenesis was significantly increased in MSC-transplanted versus non-transplanted animals. Most MSC were found in the region of the substantia nigra and the surrounding arachnoid mater, expressing S100ß and brain-derived neurotrophic factor, whereas some MSC showed an endothelial phenotype and localized around blood vessels. CONCLUSIONS: The acute neurogenic effects and neurotrophic factor expression of MSC could help to restore the SVZ-OB axis in PD.


Assuntos
Tecido Adiposo/citologia , Ventrículos Laterais/crescimento & desenvolvimento , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Neurogênese/fisiologia , Doença de Parkinson/terapia , Adulto , Animais , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Proliferação de Células/efeitos dos fármacos , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Ventrículos Laterais/citologia , Obesidade/metabolismo , Bulbo Olfatório/citologia , Oxidopamina/efeitos adversos , Ratos , Ratos Wistar , Subunidade beta da Proteína Ligante de Cálcio S100/biossíntese , Substância Negra/citologia , Adulto Jovem
12.
Eur J Immunol ; 43(3): 793-804, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23255246

RESUMO

Cytokine memory for IFN-γ production by effector/memory Th1 cells plays a key role in both protective and pathological immune responses. To understand the epigenetic mechanism determining the ontogeny of effector/memory Th1 cells characterized by stable effector functions, we identified a T-cell-specific methylation pattern at the IFNG promoter and CNS-1 in ex vivo effector/memory Th1 cells, and investigated methylation dynamics of these regions during the development of effector/memory Th1 cells. During Th1 differentiation, demethylation occurred at both the promoter and CNS-1 regions of IFNG as early as 16 h, and this process was independent of cell proliferation and DNA synthesis. Using an IFN-γ capture assay, we found early IFN-γ-producing cells from 2-day differentiating cultures acquired "permissive" levels of demethylation and developed into effector/memory Th1 cells undergoing progressive demethylation at the IFNG promoter and CNS-1 when induced by IL-12. Methylation levels of these regions in effector/memory Th1 cells of peripheral blood from rheumatoid arthritis patients correlated inversely with reduced frequencies of IFN-γ-producers, coincident with recruitment of effector/memory Th1 cells to the site of inflammation. Thus, after termination of TCR stimulation, IL-12 signaling potentiates the stable functional IFN-γ memory in effector/memory Th1 cells characterized by hypomethylation at the IFNG promoter and CNS-1.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Metilação de DNA , Memória Imunológica , Interferon gama/genética , Sequências Reguladoras de Ácido Nucleico , Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Diferenciação Celular , Células Cultivadas , Epigênese Genética , Humanos , Interferon gama/metabolismo , Interleucina-12/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais , Células Th1/citologia , Células Th1/imunologia , Células Th1/metabolismo
13.
Adv Sci (Weinh) ; 11(21): e2308447, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38491873

RESUMO

Beyond SARS-CoV2 vaccines, mRNA drugs are being explored to overcome today's greatest healthcare burdens, including cancer and cardiovascular disease. Synthetic mRNA triggers immune responses in transfected cells, which can be reduced by chemically modified nucleotides. However, the side effects of mRNA-triggered immune activation on cell function and how different nucleotides, such as the N1-methylpseudouridine (m1Ψ) used in SARS-CoV2 vaccines, can modulate cellular responses is not fully understood. Here, cellular responses toward a library of uridine-modified mRNAs are investigated in primary human cells. Targeted proteomics analyses reveal that unmodified mRNA induces a pro-inflammatory paracrine pattern marked by the secretion of chemokines, which recruit T and B lymphocytes toward transfected cells. Importantly, the magnitude of mRNA-induced changes in cell function varies quantitatively between unmodified, Ψ-, m1Ψ-, and 5moU-modified mRNA and can be gradually tailored, with implications for deliberately exploiting this effect in mRNA drug design. Indeed, both the immunosuppressive effect of stromal cells on T-cell proliferation, and the anti-inflammatory effect of IL-10 mRNA are enhanced by appropriate uridine modification. The results provide new insights into the effects of mRNA drugs on cell function and cell-cell communication and open new possibilities to tailor mRNA-triggered immune activation to the desired pro- or anti-inflammatory application.


Assuntos
RNA Mensageiro , Uridina , Humanos , Uridina/farmacologia , Uridina/imunologia , RNA Mensageiro/genética , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo , Quimiocinas/metabolismo , Quimiocinas/genética , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , COVID-19/imunologia , COVID-19/prevenção & controle , Células Cultivadas
14.
Transgenic Res ; 22(2): 251-4, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23180363

RESUMO

Here we introduce the "Tet-Transgenic Rodents" database, documenting most of the published Tet-transgenic mouse lines generated in the past 2 decades. Aside from the >500 mouse lines listed, it also includes the first of the recently reported Tet-transgenic rat models. Since the Tet technology comprises two essential components, a cis-acting promoter (Ptet) and a trans-acting transactivator, the database has been organized accordingly. One section of the database summarizes the different transgenic mouse lines carrying mostly tissue specific promoters driving the Tet transactivator. Another section covers transgenic mouse lines carrying responder transgenes under Ptet control. The few existing rat transgenic lines are listed correspondingly. It is the purpose of this database to facilitate the repeated use of preexisting, validated transgenic lines as a shortcut for further research.


Assuntos
Animais Geneticamente Modificados/classificação , Bases de Dados Genéticas , Proteínas Repressoras/genética , Animais , Animais Geneticamente Modificados/genética , Camundongos , Regiões Promotoras Genéticas , Ratos
15.
ACS Appl Mater Interfaces ; 15(37): 43219-43222, 2023 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-37676755

RESUMO

In the original paper, Li and co-workers [ACS Appl. Mater. Interfaces 2022, 14, 17128-17141] described their approach to select specific hybridoma cells from a polyclonal hybridoma pool by using a cell surface anchor to catch the secreted antibody. The antigen-specific detection was performed with streptavidin-labeled antigen and a PE-labeled anti-F(ab')2 antibody. The present comment offers a clearer description of the selection system originally published by Listek et al. in 2020 and provides further information about the importance of controls and recent adaptations made by our lab.


Assuntos
Anticorpos Monoclonais , Humanos , Animais , Hibridomas , Membrana Celular , Animais Geneticamente Modificados , Transporte Biológico
16.
Biomaterials ; 294: 121971, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36634491

RESUMO

In vitro transcribed (IVT-)mRNA has entered center stage for vaccine development due to its immune co-stimulating properties. Given the widely demonstrated safety of IVT-mRNA-based vaccines, we aimed to adopt IVT-mRNA encoding VEGF for secretory phenotype modulation of therapeutic cells. However, we observed that the immunogenicity of IVT-mRNA impairs the endogenous secretion of pro-angiogenic mediators from transfected mesenchymal stromal cells, instead inducing anti-angiogenic chemokines. This inflammatory secretome modulation limits the application potential of unmodified IVT-mRNA for cell therapy manufacturing, pro-angiogenic therapy and regenerative medicine. To uncouple immunogenicity from the protein expression functionality, we immuno-engineered IVT-mRNA with different chemically modified ribonucleotides. 5-Methoxy-uridine-modification of IVT-mRNA rescued the endogenous secretome pattern of transfected cells and prolonged secretion of IVT-mRNA-encoded VEGF. We found that high secretion of IVT-mRNA-encoded protein further depends on optimized cell adhesion. Cell encapsulation in a collagen-hyaluronic acid hydrogel increased secretion of IVT-mRNA-encoded VEGF and augmented the endogenous secretion of supporting pro-angiogenic mediators, such as HGF. Integrating minimally immunogenic mRNA technology with predesigned matrix-derived cues allows for the synergistic combination of multiple dimensions of cell manipulation and opens routes for biomaterial-based delivery of mRNA-engineered cell products. Such multimodal systems could present a more biologically relevant way to therapeutically address complex multifactorial processes such as tissue ischemia, angiogenesis, and regeneration.


Assuntos
Células-Tronco Mesenquimais , Fator A de Crescimento do Endotélio Vascular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Secretoma , Medicina Regenerativa/métodos
17.
Drug Deliv Transl Res ; 13(6): 1745-1765, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36853436

RESUMO

There is currently no causal treatment available for Parkinson's disease (PD). However, the use of glial cell line-derived neurotrophic factor (GDNF) to provide regenerative effects for neurons is promising. Such approaches require translational delivery systems that are functional in diseased tissue. To do so, we used a non-viral Sleeping Beauty (SB) transposon system to overexpress GDNF in adipose tissue-derived mesenchymal stromal cells (adMSCs). Entrapment of cells in fibrin hydrogel was used to boost potential neurorestorative effects. Functional GDNF-adMSCs were able to secrete 1066.8 ± 169.4 ng GDNF/120,000 cells in vitro. The GDNF-adMSCs were detectable for up to 1 month after transplantation in a mild 6-hydroxydopamine (6-OHDA) hemiparkinson male rat model. Entrapment of GDNF-adMSCs enabled GDNF secretion in surrounding tissue in a more concentrated manner, also tending to prolong GDNF secretion relatively. GDNF-adMSCs entrapped in hydrogel also led to positive immunomodulatory effects via an 83% reduction of regional IL-1ß levels compared to the non-entrapped GDNF-adMSC group after 1 month. Furthermore, GDNF-adMSC-treated groups showed higher recovery of tyrosine hydroxylase (TH)-expressing cells, indicating a neuroprotective function, although this was not strong enough to show significant improvement in motor performance. Our findings establish a promising GDNF treatment system in a PD model. Entrapment of GDNF-adMSCs mediated positive immunomodulatory effects. Although the durability of the hydrogel needs to be extended to unlock its full potential for motor improvements, the neuroprotective effects of GDNF were evident and safe. Further motor behavioral tests and other disease models are necessary to evaluate this treatment option adequately.


Assuntos
Células-Tronco Mesenquimais , Doença de Parkinson , Ratos , Masculino , Animais , Doença de Parkinson/etiologia , Doença de Parkinson/terapia , Ratos Sprague-Dawley , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Hidrogéis , Modelos Animais de Doenças
18.
Aging Cell ; 22(3): e13744, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36514868

RESUMO

Tissue formation and healing both require cell proliferation and migration, but also extracellular matrix production and tensioning. In addition to restricting proliferation of damaged cells, increasing evidence suggests that cellular senescence also has distinct modulatory effects during wound healing and fibrosis. Yet, a direct role of senescent cells during tissue formation beyond paracrine signaling remains unknown. We here report how individual modules of the senescence program differentially influence cell mechanics and ECM expression with relevance for tissue formation. We compared DNA damage-mediated and DNA damage-independent senescence which was achieved through over-expression of either p16Ink4a or p21Cip1 cyclin-dependent kinase inhibitors in primary human skin fibroblasts. Cellular senescence modulated focal adhesion size and composition. All senescent cells exhibited increased single cell forces which led to an increase in tissue stiffness and contraction in an in vitro 3D tissue formation model selectively for p16 and p21-overexpressing cells. The mechanical component was complemented by an altered expression profile of ECM-related genes including collagens, lysyl oxidases, and MMPs. We found that particularly the lack of collagen and lysyl oxidase expression in the case of DNA damage-mediated senescence foiled their intrinsic mechanical potential. These observations highlight the active mechanical role of cellular senescence during tissue formation as well as the need to synthesize a functional ECM network capable of transferring and storing cellular forces.


Assuntos
Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina , Humanos , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proliferação de Células , Matriz Extracelular/metabolismo
19.
Stem Cell Res Ther ; 14(1): 296, 2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-37840130

RESUMO

BACKGROUND: Direct cardiac reprogramming is currently being investigated for the generation of cells with a true cardiomyocyte (CM) phenotype. Based on the original approach of cardiac transcription factor-induced reprogramming of fibroblasts into CM-like cells, various modifications of that strategy have been developed. However, they uniformly suffer from poor reprogramming efficacy and a lack of translational tools for target cell expansion and purification. Therefore, our group has developed a unique approach to generate proliferative cells with a pre-CM phenotype that can be expanded in vitro to yield substantial cell doses. METHODS: Cardiac fibroblasts were reprogrammed toward CM fate using lentiviral transduction of cardiac transcriptions factors (GATA4, MEF2C, TBX5, and MYOCD). The resulting cellular phenotype was analyzed by RNA sequencing and immunocytology. Live target cells were purified based on intracellular CM marker expression using molecular beacon technology and fluorescence-activated cell sorting. CM commitment was assessed using 5-azacytidine-based differentiation assays and the therapeutic effect was evaluated in a mouse model of acute myocardial infarction using echocardiography and histology. The cellular secretome was analyzed using mass spectrometry. RESULTS: We found that proliferative CM precursor-like cells were part of the phenotype spectrum arising during direct reprogramming of fibroblasts toward CMs. These induced CM precursors (iCMPs) expressed CPC- and CM-specific proteins and were selectable via hairpin-shaped oligonucleotide hybridization probes targeting Myh6/7-mRNA-expressing cells. After purification, iCMPs were capable of extensive expansion, with preserved phenotype when under ascorbic acid supplementation, and gave rise to CM-like cells with organized sarcomeres in differentiation assays. When transplanted into infarcted mouse hearts, iCMPs prevented CM loss, attenuated fibrotic scarring, and preserved ventricular function, which can in part be attributed to their substantial secretion of factors with documented beneficial effect on cardiac repair. CONCLUSIONS: Fibroblast reprogramming combined with molecular beacon-based cell selection yields an iCMP-like cell population with cardioprotective potential. Further studies are needed to elucidate mechanism-of-action and translational potential.


Assuntos
Infarto do Miocárdio , Miócitos Cardíacos , Camundongos , Animais , Miócitos Cardíacos/metabolismo , Remodelação Ventricular , Proteínas com Domínio T/genética , Fatores de Transcrição MEF2/genética , Infarto do Miocárdio/terapia , Infarto do Miocárdio/tratamento farmacológico , Fibroblastos , Reprogramação Celular/genética
20.
Mol Ther Nucleic Acids ; 34: 102068, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38034031

RESUMO

Dysfunction of endothelial cells (ECs) lining the inner surface of blood vessels are causative for a number of diseases. Hence, the ability to therapeutically modulate gene expression within ECs is of high therapeutic value in treating diseases such as those associated with lung edema. mRNAs formulated with lipid nanoparticles (LNPs) have emerged as a new drug modality to induce transient protein expression for modulating disease-relevant signal transduction pathways. In the study presented here, we tested the effect of a novel synthetic, nucleoside-modified mRNA encoding COMP-Ang1 (mRNA-76) formulated into a cationic LNP on attenuating inflammation-induced vascular leakage. After intravenous injection, the respective mRNA was found to be delivered almost exclusively to the ECs of the lung, while sparing other vascular beds and bypassing the liver. The mode of action of mRNA-76, such as its activation of the Tie2 signal transduction pathway, was tested by pharmacological studies in vitro and in vivo in respective mouse models. mRNA-76 was found to prevent lung vascular leakage/lung edema as well as neutrophil infiltration in a lipopolysaccharide-challenging model.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA