Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 7(1): 865, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39009807

RESUMO

Long-acting passive immunization strategies are needed to protect immunosuppressed vulnerable groups from infectious diseases. To further explore this concept for COVID-19, we constructed Adeno-associated viral (AAV) vectors encoding the human variable regions of the SARS-CoV-2 neutralizing antibody, TRES6, fused to murine constant regions. An optimized vector construct was packaged in hepatotropic (AAV8) or myotropic (AAVMYO) AAV capsids and injected intravenously into syngeneic TRIANNI-mice. The highest TRES6 serum concentrations (511 µg/ml) were detected 24 weeks after injection of the myotropic vector particles and mean TRES6 serum concentrations remained above 100 µg/ml for at least one year. Anti-drug antibodies or TRES6-specific T cells were not detectable. After injection of the AAV8 particles, vector mRNA was detected in the liver, while the AAVMYO particles led to high vector mRNA levels in the heart and skeletal muscle. The analysis of the Fc-glycosylation pattern of the TRES6 serum antibodies revealed critical differences between the capsids that coincided with different binding activities to murine Fc-γ-receptors. Concomitantly, the vector-based immune prophylaxis led to protection against SARS-CoV-2 infection in K18-hACE2 mice. High and long-lasting expression levels, absence of anti-drug antibodies and favourable Fc-γ-receptor binding activities warrant further exploration of myotropic AAV vector-based delivery of antibodies and other biologicals.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19 , Dependovirus , Vetores Genéticos , Receptores de IgG , SARS-CoV-2 , Animais , Dependovirus/genética , SARS-CoV-2/imunologia , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Camundongos , Humanos , COVID-19/imunologia , COVID-19/prevenção & controle , Vetores Genéticos/genética , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Receptores de IgG/metabolismo , Receptores de IgG/genética , Receptores de IgG/imunologia , Tropismo Viral , Imunização Passiva
2.
Adv Mater ; 36(28): e2403642, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38653478

RESUMO

Myocardial infarction (MI) causes cell death, disrupts electrical activity, triggers arrhythmia, and results in heart failure, whereby 50-60% of MI-associated deaths manifest as sudden cardiac deaths (SCD). The most effective therapy for SCD prevention is implantable cardioverter defibrillators (ICDs). However, ICDs contribute to adverse remodeling and disease progression and do not prevent arrhythmia. This work develops an injectable collagen-PEDOT:PSS (poly(3,4-ethylenedioxythiophene) polystyrene sulfonate) hydrogel that protects infarcted hearts against ventricular tachycardia (VT) and can be combined with human induced pluripotent stem cell (hiPSC)-cardiomyocytes to promote partial cardiac remuscularization. PEDOT:PSS improves collagen gel formation, micromorphology, and conductivity. hiPSC-cardiomyocytes in collagen-PEDOT:PSS hydrogels exhibit near-adult sarcomeric length, improved contractility, enhanced calcium handling, and conduction velocity. RNA-sequencing data indicate enhanced maturation and improved cell-matrix interactions. Injecting collagen-PEDOT:PSS hydrogels in infarcted mouse hearts decreases VT to the levels of healthy hearts. Collectively, collagen-PEDOT:PSS hydrogels offer a versatile platform for treating cardiac injuries.


Assuntos
Arritmias Cardíacas , Colágeno , Condutividade Elétrica , Hidrogéis , Células-Tronco Pluripotentes Induzidas , Infarto do Miocárdio , Miócitos Cardíacos , Poliestirenos , Infarto do Miocárdio/patologia , Animais , Células-Tronco Pluripotentes Induzidas/citologia , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Camundongos , Colágeno/química , Hidrogéis/química , Arritmias Cardíacas/prevenção & controle , Poliestirenos/química , Polímeros/química , Compostos Bicíclicos Heterocíclicos com Pontes/química , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Tiofenos
3.
J Mater Chem B ; 12(15): 3774-3785, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38535706

RESUMO

The regeneration of bone defects that exceed 2 cm is a challenge for the human body, necessitating interventional therapies. Demineralized bone matrices (DBM) derived from biological tissues have been employed for bone regeneration and possess notable osteoinductive and osteoconductive characteristics. Nevertheless, their efficiency in regenerating critically sized injuries is limited, and therefore additional signaling cues are required. Thanks to the piezoelectric properties of the bone, external physical stimulation is shown to accelerate tissue healing. We have implanted human DBM in critically sized cranial bone defects in rat animal models and exposed them to an external magnetic field (1 T) to enhance endogenous bone formation. Our in vitro experiments showed the superior cytocompatibility of DBM compared to cell culture plates. Furthermore, alkaline phosphatase activity after 14 days and Alizarin red staining at 28 days demonstrated differentiation of rat bone marrow mesenchymal stem cells into bone lineage on DBM. Computer tomography images together with histological analyses showed that implanting DBM in the injured rats significantly enhanced bone regeneration. Notably, combining DBM transplantation with a 2 h daily exposure to a 1 T magnetic field for 2 weeks (day 7 to 21 post-surgery) significantly improved bone regeneration compared to DBM transplantation alone. This research indicates that utilizing external magnetic stimulation significantly enhances the potential of bone allografts to regenerate critically sized bone defects.


Assuntos
Matriz Óssea , Osso e Ossos , Ratos , Humanos , Animais , Regeneração Óssea , Osteogênese , Modelos Animais
4.
J Mater Chem B ; 11(31): 7280-7299, 2023 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-37427687

RESUMO

Cardiovascular diseases are the primary cause of death worldwide. Despite significant advances in pharmacological treatments and surgical interventions to restore heart function after myocardial infarction, it can progress to heart failure due to the restricted inherent potential of adult cardiomyocytes to self-regenerate. Hence, the evolution of new therapeutic methods is critical. Nowadays, novel approaches in tissue engineering have assisted in restoring biological and physical specifications of the injured myocardium and, hence, cardiac function. The incorporation of a supporting matrix that could mechanically and electronically support the heart tissue and stimulate the cells to proliferate and regenerate will be advantageous. Electroconductive nanomaterials can facilitate intracellular communication and aid synchronous contraction via electroactive substrate creation, preventing the issue of arrhythmia in the heart. Among a wide range of electroconductive materials, graphene-based nanomaterials (GBNs) are promising for cardiac tissue engineering (CTE) due to their outstanding features including high mechanical strength, angiogenesis, antibacterial and antioxidant properties, low cost, and scalable fabrication. In the present review, we discuss the effect of applying GBNs on angiogenesis, proliferation, and differentiation of implanted stem cells, their antibacterial and antioxidant properties, and their role in improving the electrical and mechanical properties of the scaffolds for CTE. Also, we summarize the recent research that has applied GBNs in CTE. Finally, we present a concise discussion on the challenges and prospects.


Assuntos
Grafite , Nanoestruturas , Engenharia Tecidual , Grafite/farmacologia , Grafite/química , Antioxidantes , Antibacterianos
5.
Adv Healthc Mater ; 12(20): e2202408, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36976709

RESUMO

Cardiac tissue engineering is a promising strategy to prevent heart failure. However, several issues remain unsolved, including efficient electrical coupling and incorporating factors to enhance tissue maturation and vascularization. Herein, a biohybrid hydrogel that enhances beating properties of engineered cardiac tissues and allows drug release concurrently is developed. Gold nanoparticles (AuNPs) with different sizes (18-241 nm) and surface charges (33.9-55.4 mV) are synthesized by reducing gold (III) chloride trihydrate using branched polyethyleneimine (bPEI). These nanoparticles increase gel stiffness from ≈91 to ≈146 kPa, enhance electrical conductivity of collagen hydrogels from ≈40 to 49-68 mS cm-1 , and allow slow and steady release of loaded drugs. Engineered cardiac tissues based on bPEI-AuNP-collagen hydrogels and either primary or human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes show enhanced beating properties. hiPSC-derived cardiomyocytes exhibit more aligned and wider sarcomeres in bPEI-AuNP-collagen hydrogels compared to collagen hydrogels. Furthermore, the presence of bPEI-AuNPs result in advanced electrical coupling evidenced by synchronous and homogenous calcium flux throughout the tissue. RNA-seq analyses are in agreement with these observations. Collectively, this data demonstrate the potential of bPEI-AuNP-collagen hydrogels to improve tissue engineering approaches to prevent heart failure and possibly treat diseases of other electrically sensitive tissues.


Assuntos
Insuficiência Cardíaca , Células-Tronco Pluripotentes Induzidas , Nanopartículas Metálicas , Humanos , Ouro , Engenharia Tecidual , Polietilenoimina , Hidrogéis/farmacologia , Liberação Controlada de Fármacos , Miócitos Cardíacos , Colágeno
6.
Biofabrication ; 15(1)2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36279872

RESUMO

Type 1 diabetes mellitus is an autoimmune disease characterized by the loss of pancreatic isletßcells. Insulin injections and pancreas transplants are currently available therapies. The former requires daily insulin injections, while the latter is constrained by donor organ availability. Islet transplantation is a promising alternative treatment for type 1 diabetes mellitus that may overcome the limitations of previous techniques. Two challenges, however, must be addressed: limited cell retention as a result of the immune response and limited function of the transplanted cells that survive. To address these problems, we developed a microfluidic technology for a one-step generation of islet-laden fibers to protect them from the immune response. This approach enables continuous generation of microfibers with a diameter suitable for islet encapsulation (275µm). We, then, transplanted islet-laden fibers into diabetic Wistar rats. While islet-laden fibers alone were unable to restore normoglycemia in diabetic rats, adding mesenchymal stromal cells (MSCs) restored normoglycemia for an extended time. It increased the animals' lifespan by up to 75 d. Additionally, it improved the glucose-stimulated response of islets to the point where there was no significant difference between the treatment group and the healthy animals. Additionally, the presence of MSCs suppressed the immune response, as seen by decreased levels of pro-inflammatory cytokines such as tumor necrosis factor-α. Taken together, these fibers including islet and MSCs provide a versatile platform for concurrently improving cell preservation and functioning followingin vivotransplantation.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Células-Tronco Mesenquimais , Ratos , Animais , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/patologia , Longevidade , Ratos Wistar , Transplante das Ilhotas Pancreáticas/métodos , Células-Tronco Mesenquimais/fisiologia , Insulina
7.
ACS Biomater Sci Eng ; 8(3): 1258-1270, 2022 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35193354

RESUMO

While bone regenerates itself after an injury, a critical bone defect requires external interventions. Engineering approaches to restore bone provide a temporary scaffold to support the damage and provide beneficial biological cues for bone repair. Biomimetically generated scaffolds replicate the naturally occurring phenomena in bone regeneration. In this study, a gelatin-calcium phosphate nanocomposite was synthesized by an efficient and cost-effective double-diffusion biomimetic approach. Calcium and phosphate ions are impregnated in the gelatin, mimicking the natural bone mineralization process. Glutaraldehyde from 0.5 to 2 w/v% was used for gelatin cross-linking and mechanical properties of the scaffold, and its biological support for rat bone marrow mesenchymal stromal cells was analyzed. Analysis of scanning electron microscopy images of the nanocomposite scaffolds and Fourier transform infrared (FTIR) and X-ray diffraction (XRD) characterizations of these scaffolds confirmed precipitation of calcium phosphates in the gelatin. Moreover, lysozyme degradation assay showed that scaffold degradation reversely correlates with the concentration of the cross-linking agent. Increased glutaraldehyde concentrations enhanced the mechanical properties of the scaffolds, bringing them closer to those of cancellous bone. Rat bone marrow mesenchymal stromal cells maintained their viability on these scaffolds compared to standard cell culture plates. In addition, these cells showed differentiation into bone lineage as evaluated from alkaline phosphatase activity up to 21 days and Alizarin red staining of the cells over 28 days. Eventually, scaffolds were implanted in a cranial defect in a rat animal model with a 5 mm diameter. Bone regeneration was studied over 90 days. Analysis of histological sections of the injury and computer tomography images revealed that nanocomposite scaffolds cross-linked with 1% w/v glutaraldehyde provide the maximum bone regeneration after 90 days. Collectively, our data show that nanocomposite scaffolds developed here provide effective regeneration for extensive bone defects in vivo.


Assuntos
Gelatina , Nanocompostos , Animais , Biomimética , Gelatina/farmacologia , Glutaral/farmacologia , Modelos Animais , Ratos , Engenharia Tecidual/métodos , Alicerces Teciduais
8.
Adv Healthc Mater ; 10(20): e2100926, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34499814

RESUMO

Cardiac tissue engineering is a promising strategy to generate human cardiac tissues for modeling cardiac diseases, screening for therapeutic drugs, and repairing the injured heart. Yet, several issues remain to be resolved including the generation of tissues with high cardiomyocyte density. Here, it is shown that the integration of the glycogen synthase kinase-3ß inhibitor CHIR99021 in collagen I hydrogels promotes proliferation of human-induced pluripotent stem cell-derived (hiPSC) cardiomyocytes post-fabrication improving contractility of and calcium flow in engineered 3D cardiac microtissues. CHIR99021 has no effect on the gelation kinetics or the mechanical properties of collagen I hydrogels. Analysis of cell density and proliferation based on Ki-67 staining indicates that integration of CHIR99021 together with external CHIR99021 stimulation increases hiPSC-cardiomyocyte number by ≈2-fold within 7 d post-fabrication. Analysis of the contractility of engineered cardiac tissues after another 3 d in the absence of external CHIR99021 shows that CHIR99021-induced hiPSC-cardiomyocyte proliferation results in synchronized calcium flow, rhythmic beating, increased speed of contraction and contraction amplitude, and reduced peak-to-peak time. The CHIR99021-stimulated engineered cardiac microtissues exhibit spontaneous rhythmic contractions for at least 35 d. Collectively, the data demonstrate the potential of induced cardiomyocyte proliferation to enhance engineered cardiac microtissues by increasing cardiomyocyte density.


Assuntos
Células-Tronco Pluripotentes Induzidas , Diferenciação Celular , Proliferação de Células , Humanos , Miócitos Cardíacos , Piridinas/farmacologia , Pirimidinas
9.
Antioxid Redox Signal ; 35(3): 143-162, 2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32993354

RESUMO

Significance: Heart failure is among the leading causes of morbidity worldwide with a 5-year mortality rate of ∼50%. Therefore, major efforts are invested to reduce heart damage upon injury or maintain and at best restore heart function. Recent Advances: In clinical trials, acellular constructs succeeded in improving cardiac function by stabilizing the infarcted heart. In addition, strategies utilizing stem-cell-derived cardiomyocytes have been developed to improve heart function postmyocardial infarction in small and large animal models. These strategies range from injection of cell-laden hydrogels to unstructured hydrogel-based and complex biofabricated cardiac patches. Importantly, novel methods have been developed to promote differentiation of stem-cell-derived cardiomyocytes to prevascularized cardiac patches. Critical Issues: Despite substantial progress in vascularization strategies for heart-on-the-chip technologies, little advance has been made in generating vascularized cardiac patches with clinically relevant dimensions. In addition, proper electrical coupling between engineered and host tissue to prevent and/or eliminate arrhythmia remains an unresolved issue. Finally, despite advanced approaches to include hierarchical structures in cardiac tissues, engineered tissues do not generate forces in the range of native adult cardiac tissue. Future Directions: It involves utilizing novel materials and advancing biofabrication strategies to generate prevascularized three-dimensional multicellular constructs of clinical relevant size; inclusion of hierarchical structures, electroconductive materials, and biologically active factors to enhance cardiomyocyte differentiation for optimized force generation and vascularization; optimization of bioreactor strategies for tissue maturation. Antioxid. Redox Signal. 35, 143-162.


Assuntos
Miócitos Cardíacos/metabolismo , Medicina Regenerativa , Células-Tronco/metabolismo , Engenharia Tecidual , Animais , Materiais Biocompatíveis , Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Modelos Animais de Doenças , Fenômenos Eletrofisiológicos , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/terapia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Miócitos Cardíacos/citologia , Neovascularização Fisiológica , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais
10.
Sci Rep ; 10(1): 8789, 2020 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-32472031

RESUMO

Cardiac tissue engineering is a promising approach to treat cardiovascular diseases, which are a major socio-economic burden worldwide. An optimal material for cardiac tissue engineering, allowing cardiomyocyte attachment and exhibiting proper immunocompatibility, biocompatibility and mechanical characteristics, has not yet emerged. An additional challenge is to develop a fabrication method that enables the generation of proper hierarchical structures and constructs with a high density of cardiomyocytes for optimal contractility. Thus, there is a focus on identifying suitable materials for cardiac tissue engineering. Here, we investigated the interaction of neonatal rat heart cells with engineered spider silk protein (eADF4(C16)) tagged with the tripeptide arginyl-glycyl-aspartic acid cell adhesion motif RGD, which can be used as coating, but can also be 3D printed. Cardiomyocytes, fibroblasts, and endothelial cells attached well to eADF4(C16)-RGD coatings, which did not induce hypertrophy in cardiomyocytes, but allowed response to hypertrophic as well as proliferative stimuli. Furthermore, Kymograph and MUSCLEMOTION analyses showed proper cardiomyocyte beating characteristics on spider silk coatings, and cardiomyocytes formed compact cell aggregates, exhibiting markedly higher speed of contraction than cardiomyocyte mono-layers on fibronectin. The results suggest that eADF4(C16)-RGD is a promising material for cardiac tissue engineering.


Assuntos
Fibroínas/química , Miócitos Cardíacos/citologia , Oligopeptídeos/química , Engenharia Tecidual/métodos , Animais , Animais Recém-Nascidos , Materiais Biocompatíveis/química , Adesão Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ratos , Proteínas Recombinantes/química
11.
Biomater Sci ; 7(9): 3906-3917, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31322163

RESUMO

Cardiovascular diseases represent a major socio-economic burden. In recent years, considerable effort has been invested in optimizing cell delivery strategies to advance cell transplantation therapies to restore heart function for example after an infarct. A particular issue is that the implantation of cells using a non-electroconductive matrix potentially causes arrhythmia. Here, we demonstrate that our hydrazide-functionalized nanotubes-pericardial matrix-derived electroconductive biohybrid hydrogel provides a suitable environment for maturation of human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes. hiPSC-derived cardiomyocytes exhibited an improved contraction amplitude (>500%) on conductive hydrogels compared to cells cultured on Matrigel®. This was accompanied by increased cellular alignment, enhanced connexin 43 expression, and improved sarcomere organization suggesting maturation of the hiPSC-derived cardiomyocytes. Sarcomeric length of these cells increased from 1.3 to 1.7 µm. Moreover, 3D cell-laden engineered tissues exhibited enhanced calcium handling as well as positive response to external electrical and pharmaceutical stimulation. Collectively, our data indicate that our biohybrid hydrogels consisting of solubilized nanostructured pericardial matrix and electroconductive positively charged hydrazide-conjugated carbon nanotubes provide a promising material for stem cell-based cardiac tissue engineering.


Assuntos
Materiais Biocompatíveis/química , Hidrogéis/química , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Nanotubos de Carbono/química , Pericárdio/química , Alicerces Teciduais/química , Biomarcadores/metabolismo , Cálcio/metabolismo , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Colágeno/química , Conexina 43/metabolismo , Combinação de Medicamentos , Condutividade Elétrica , Humanos , Laminina/química , Células-Tronco Mesenquimais/citologia , Tamanho da Partícula , Proteoglicanas/química
12.
Expert Opin Biol Ther ; 19(2): 105-118, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30570406

RESUMO

INTRODUCTION: Vascularization remains one of the greatest yet unmet challenges in tissue engineering. When engineered tissues are scaled up to therapeutically relevant dimensions, their demand of oxygen and nutrients can no longer be met by diffusion. Thus, there is a need for perfusable vascular structures. Hypoxia-inducible factors (HIF) act as transcriptional oxygen sensors and regulate a multitude of genes involved in adaptive processes to hypoxia, including angiogenesis. Thus, targeting HIFs is a promising strategy to induce vascularization of engineered tissues. AREAS COVERED: Here we review current vascularization strategies and summarize the present knowledge regarding activation of HIF signaling by ions, iron chelating agents, α-Ketoglutarate (αKG) analogues, and the lipid-lowering drug simvastatin to induce angiogenesis. Specifically, we focus on the incorporation of HIF-activating agents into biomaterials and scaffolds for controlled release. EXPERT OPINION: Vascularization of tissue constructs through activation of upstream regulators of angiogenesis offers advantages but also suffers from drawbacks. HIFs can induce a complete angiogenic program; however, this program appears to be too slow to vascularize larger constructs before cell death occurs. It is therefore crucial that HIF-activation is combined with cell protective strategies and prevascularization techniques to obtain fully vascularized, vital tissues of therapeutically relevant dimensions.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Neovascularização Fisiológica , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Humanos , Fator 1 Induzível por Hipóxia/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA