Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
1.
Sci Rep ; 12(1): 1227, 2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35075209

RESUMO

Cirrhosis refers to irreversible liver damage where healthy tissue is replaced by scar tissue, resulting in impaired liver function. There is no cure and current treatments only prevent further liver damage; thus, novel therapeutic options are urgently needed. Here, we report a new approach that enables the formation of self-assembled 3D spheroids of adipose-derived stem cells (ADSCs) and murine hepatocytes (AML12) via reconstituted collagen fibers. Compared with the spheroids formed in the commercially available EZSHERE dish, the collagen fiber-based ADSC/hepatocyte spheroids offer a notable benefit in structure formation and paracrine factor secretion. To test the regenerative capability of the collagen fiber-based 3D ADSC/hepatocyte spheroids, a rat model of thioacetamide (TAA)-induced liver cirrhosis was employed. The transplantation of the collagen fiber-based 3D ADSC/hepatocyte spheroids show an improvement in liver function and ameliorates pathological liver cirrhosis in TAA-treated rats. In summary, our data show collagen fiber-based self-assembled 3D ADSC/hepatocyte spheroids to possess the excellent regenerative capacity in response to TAA-induced liver injury, promising an alternative therapeutic strategy for liver cirrhosis.


Assuntos
Hepatócitos/transplante , Cirrose Hepática/terapia , Esferoides Celulares/transplante , Animais , Colágeno/metabolismo , Modelos Animais de Doenças , Humanos , Cirrose Hepática/induzido quimicamente , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Ratos Sprague-Dawley , Tioacetamida
3.
J Tissue Eng Regen Med ; 15(12): 1131-1143, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34551191

RESUMO

Volumetric muscle loss (VML) is traumatic or surgical loss of skeletal muscle with resultant functional impairment. Skeletal muscle's innate capacity for regeneration is lost with VML due to a critical loss of stem cells, extracellular matrix, and neuromuscular junctions. Consequences of VML include permanent disability or delayed amputations of the affected limb. Currently, a successful clinical therapy has not been identified. Mesenchymal stem cells (MSCs) possess regenerative and immunomodulatory properties and their three-dimensional aggregation can further enhance therapeutic efficacy. In this study, MSC aggregation into spheroids was optimized in vitro based on cellular viability, spheroid size, and trophic factor secretion. The regenerative potential of the optimized MSC spheroid therapy was then investigated in a murine model of VML injury. Experimental groups included an untreated VML injury control, intramuscular injection of MSC spheroids, and MSC spheroids encapsulated in a fibrin-laminin hydrogel. Compared to the untreated VML group, the spheroid encapsulating hydrogel group enhanced myogenic marker (i.e., MyoD and myogenin) protein expression, improved muscle mass, increased presence of centrally nucleated myofibers as well as small fibers (<500 µm2 ), modulated pro- and anti-inflammatory macrophage marker expression (i.e., iNOS and Arginase), and increased the presence of CD146+ pericytes and CD31+ endothelial cells in the VML injured muscles. Future studies will evaluate the extent of functional recovery with the spheroid encapsulating hydrogel therapy.


Assuntos
Células Imobilizadas , Fibrina/química , Hidrogéis/química , Laminina/química , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Músculo Esquelético , Regeneração , Esferoides Celulares , Ferimentos e Lesões , Animais , Células Imobilizadas/metabolismo , Células Imobilizadas/transplante , Masculino , Camundongos , Músculo Esquelético/lesões , Músculo Esquelético/fisiologia , Esferoides Celulares/metabolismo , Esferoides Celulares/transplante , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/terapia
4.
Exp Biol Med (Maywood) ; 246(22): 2372-2380, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34102903

RESUMO

Drug resistance is a major barrier against successful treatments of cancer patients. Various intrinsic mechanisms and adaptive responses of tumor cells to cancer drugs often lead to failure of treatments and tumor relapse. Understanding mechanisms of cancer drug resistance is critical to develop effective treatments with sustained anti-tumor effects. Three-dimensional cultures of cancer cells known as spheroids present a biologically relevant model of avascular tumors and have been increasingly incorporated in tumor biology and cancer drug discovery studies. In this review, we discuss several recent studies from our group that utilized colorectal tumor spheroids to investigate responses of cancer cells to cytotoxic and molecularly targeted drugs and uncover mechanisms of drug resistance. We highlight our findings from both short-term, one-time treatments and long-term, cyclic treatments of tumor spheroids and discuss mechanisms of adaptation of cancer cells to the treatments. Guided by mechanisms of resistance, we demonstrate the feasibility of designing specific drug combinations to effectively block growth and resistance of cancer cells in spheroid cultures. Finally, we conclude with our perspectives on the utility of three-dimensional tumor models and their shortcomings and advantages for phenotypic and mechanistic studies of cancer drug resistance.


Assuntos
Neoplasias Colorretais/terapia , Resistencia a Medicamentos Antineoplásicos , Esferoides Celulares/transplante , Células Tumorais Cultivadas/transplante , Animais , Antineoplásicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Humanos
5.
Sci Robot ; 6(52)2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-34043547

RESUMO

High-precision delivery of microrobots at the whole-body scale is of considerable importance for efforts toward targeted therapeutic intervention. However, vision-based control of microrobots, to deep and narrow spaces inside the body, remains a challenge. Here, we report a soft and resilient magnetic cell microrobot with high biocompatibility that can interface with the human body and adapt to the complex surroundings while navigating inside the body. We achieve time-efficient delivery of soft microrobots using an integrated platform called endoscopy-assisted magnetic actuation with dual imaging system (EMADIS). EMADIS enables rapid deployment across multiple organ/tissue barriers at the whole-body scale and high-precision delivery of soft and biohybrid microrobots in real time to tiny regions with depth up to meter scale through natural orifice, which are commonly inaccessible and even invisible by conventional endoscope and medical robots. The precise delivery of magnetic stem cell spheroid microrobots (MSCSMs) by the EMADIS transesophageal into the bile duct with a total distance of about 100 centimeters can be completed within 8 minutes. The integration strategy offers a full clinical imaging technique-based therapeutic/intervention system, which broadens the accessibility of hitherto hard-to-access regions, by means of soft microrobots.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Endoscopia/instrumentação , Robótica/instrumentação , Células 3T3 , Animais , Sistemas Computacionais , Diagnóstico por Imagem/instrumentação , Desenho de Equipamento , Feminino , Humanos , Magnetismo/instrumentação , Masculino , Camundongos , Microscopia Eletrônica de Varredura , Microtecnologia , Cirurgia Endoscópica por Orifício Natural/instrumentação , Ratos , Ratos Sprague-Dawley , Esferoides Celulares/transplante , Sus scrofa , Ultrassonografia
6.
Int J Mol Sci ; 22(8)2021 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-33923671

RESUMO

Stem cell therapy is one of the most promising candidate treatments for spinal cord injury. Research has shown optimistic results for this therapy, but clinical limitations remain, including poor viability, engraftment, and differentiation. Here, we isolated novel peripheral nerve-derived stem cells (PNSCs) from adult peripheral nerves with similar characteristics to neural-crest stem cells. These PNSCs expressed neural-crest specific markers and showed multilineage differentiation potential into Schwann cells, neuroglia, neurons, and mesodermal cells. In addition, PNSCs showed therapeutic potential by releasing the neurotrophic factors, including glial cell-line-derived neurotrophic factor, insulin-like growth factor, nerve growth factor, and neurotrophin-3. PNSC abilities were also enhanced by their development into spheroids which secreted neurotrophic factors several times more than non-spheroid PNSCs and expressed several types of extra cellular matrix. These features suggest that the potential for these PNSC spheroids can overcome their limitations. In an animal spinal cord injury (SCI) model, these PNSC spheroids induced functional recovery and neuronal regeneration. These PNSC spheroids also reduced the neuropathic pain which accompanies SCI after remyelination. These PNSC spheroids may represent a new therapeutic approach for patients suffering from SCI.


Assuntos
Esferoides Celulares/transplante , Traumatismos da Medula Espinal/terapia , Regeneração da Medula Espinal , Transplante de Células-Tronco/métodos , Animais , Células Cultivadas , Células-Tronco Neurais/citologia , Neurogênese , Nervos Periféricos/citologia , Ratos , Ratos Sprague-Dawley , Células de Schwann/citologia , Esferoides Celulares/citologia
7.
J Mol Med (Berl) ; 99(3): 425-438, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33484281

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumorigenic virus and the etiological agent of an endothelial tumor (Kaposi's sarcoma) and two B cell proliferative diseases (primary effusion lymphoma and multicentric Castleman's disease). While in patients with late stage of Kaposi's sarcoma the majority of spindle cells are KSHV-infected, viral copies are rapidly lost in vitro, both upon culture of tumor-derived cells or from newly infected endothelial cells. We addressed this discrepancy by investigating a KSHV-infected endothelial cell line in various culture conditions and in tumors of xenografted mice. We show that, in contrast to two-dimensional endothelial cell cultures, KSHV genomes are maintained under 3D cell culture conditions and in vivo. Additionally, an increased rate of newly infected cells was detected in 3D cell culture. Furthermore, we show that the PI3K/Akt/mTOR and ATM/γH2AX pathways are modulated and support an improved KSHV persistence in 3D cell culture. These mechanisms may contribute to the persistence of KSHV in tumor tissue in vivo and provide a novel target for KS specific therapeutic interventions. KEY MESSAGES: In vivo maintenance of episomal KSHV can be mimicked in 3D spheroid cultures 3D maintenance of KSHV is associated with an increased de novo infection frequency PI3K/Akt/mTOR and ATM/ γH2AX pathways contribute to viral maintenance.


Assuntos
Técnicas de Cultura de Células em Três Dimensões , Células Endoteliais/virologia , Herpesvirus Humano 8/fisiologia , Cultura de Vírus/métodos , Animais , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Transformada , Doxiciclina/farmacologia , Células Endoteliais/citologia , Genoma Viral , Xenoenxertos , Histonas/fisiologia , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Plasmídeos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Sarcoma de Kaposi/virologia , Transdução de Sinais/fisiologia , Esferoides Celulares/transplante , Esferoides Celulares/virologia , Serina-Treonina Quinases TOR/fisiologia , Latência Viral , Liberação de Vírus , Replicação Viral
8.
Bull Exp Biol Med ; 169(4): 539-543, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32910386

RESUMO

In experiments on rats, co-transplantation of olfactory ensheathing cells of the human olfactory mucosa and neural stem/progenitor cells from the same source into post-traumatic cysts of the spinal cord led to improvement of the motor activity of the hind limbs and reduced the size of the cysts in some animals by 4-12%. The transplantation of a combination of the olfactory mucosa cells is effective and can be used in preclinical trials for the treatment of spinal cord injuries.


Assuntos
Cistos/terapia , Células Epiteliais/transplante , Células-Tronco Neurais/transplante , Recuperação de Função Fisiológica/fisiologia , Esferoides Celulares/transplante , Traumatismos da Medula Espinal/terapia , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Cistos/patologia , Cistos/fisiopatologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Feminino , Humanos , Atividade Motora/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Mucosa Olfatória/citologia , Mucosa Olfatória/fisiologia , Cultura Primária de Células , Ratos , Ratos Wistar , Esferoides Celulares/citologia , Esferoides Celulares/fisiologia , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Transplante Heterólogo , Resultado do Tratamento
9.
Bull Exp Biol Med ; 169(4): 549-557, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32910392

RESUMO

Regenerative potential of multipotent mesenchymal stromal cells from the human umbilical cord (MMSC-UC) in the suspension and spheroid form was revealed during the progression of experimental small focal myocardial infarction in rats. In isoproterenol-induced myocardial infarction, foci of necrosis and inflammatory infiltrate and at later terms fibrosis foci were found mainly in the left ventricle of rat heart. In rats receiving MMSC-UC, destructive changes in the myocardium, fibrous scars, and inflammatory process were less pronounced. MMSC-UC also contributed to normalization of the morphofunctional parameters of the heart. Spheroids exhibited higher efficiency in comparison with cell suspension.


Assuntos
Fibrose Endomiocárdica/prevenção & controle , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Infarto do Miocárdio/terapia , Regeneração/fisiologia , Esferoides Celulares/transplante , Animais , Modelos Animais de Doenças , Fibrose Endomiocárdica/induzido quimicamente , Fibrose Endomiocárdica/patologia , Fibrose Endomiocárdica/fisiopatologia , Ventrículos do Coração/patologia , Ventrículos do Coração/ultraestrutura , Humanos , Isoproterenol/administração & dosagem , Masculino , Células-Tronco Mesenquimais/citologia , Infarto do Miocárdio/induzido quimicamente , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Miocárdio/ultraestrutura , Miócitos Cardíacos/patologia , Miócitos Cardíacos/ultraestrutura , Cultura Primária de Células , Ratos , Ratos Wistar , Esferoides Celulares/citologia , Esferoides Celulares/fisiologia , Transplante Heterólogo , Resultado do Tratamento , Cordão Umbilical/citologia , Cordão Umbilical/metabolismo
10.
Nanomedicine ; 29: 102253, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32619705

RESUMO

Functional articular cartilage regeneration remains challenging, and it is essential to restore focal osteochondral defects and prevent secondary osteoarthritis. Combining autologous stem cells with therapeutic medical device, we developed a bi-compartmented implant that could promote both articular cartilage and subchondral bone regeneration. The first compartment based on therapeutic collagen associated with bone morphogenetic protein 2, provides structural support and promotes subchondral bone regeneration. The second compartment contains bone marrow-derived mesenchymal stem cell spheroids to support the regeneration of the articular cartilage. Six-month post-implantation, the regenerated articular cartilage surface was 3 times larger than that of untreated animals, and the regeneration of the osteochondral tissue occurred during the formation of hyaline-like cartilage. Our results demonstrate the positive impact of this combined advanced therapy medicinal product, meeting the needs of promising osteochondral regeneration in critical size articular defects in a large animal model combining not only therapeutic implant but also stem cells.


Assuntos
Cartilagem Articular/crescimento & desenvolvimento , Transplante de Células-Tronco Mesenquimais , Osteocondrose/terapia , Próteses e Implantes , Regeneração/genética , Animais , Proteína Morfogenética Óssea 2/genética , Regeneração Óssea/genética , Regeneração Óssea/fisiologia , Cartilagem Articular/patologia , Colágeno/genética , Colágeno/farmacologia , Modelos Animais de Doenças , Humanos , Osteocondrose/genética , Osteocondrose/patologia , Ovinos/genética , Ovinos/fisiologia , Esferoides Celulares/citologia , Esferoides Celulares/transplante , Engenharia Tecidual/métodos
11.
Theranostics ; 10(16): 7409-7421, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32642002

RESUMO

Hertwig's epithelial root sheath (HERS) plays indispensable roles in tooth root development, including controlling the shape and number of roots, dentin formation, and helping generate the cementum. Based on these characteristics, HERS cell is a potential seed cell type for tooth-related tissue regeneration. However, the application is severely limited by a lack of appropriate culture methods and small cell numbers. Methods: Here, we constructed a 3D culture method to expand functional HERS cells into spheroids, and investigated characteristics and application of dental tissue regeneration of these spheroids. HERS spheroids and HERS cells (2D monolayer culture) were compared in terms of biological characteristics (such as proliferation, self-renewal capacity, and stemness) in vitro and functions (including differentiation potential and inductive ability of dentin formation) both in vitro and in vivo. Further, transcriptome analysis was utilized to reveal the molecular mechanisms of their obvious differences. Results: HERS spheroids showed obvious superiority in biological characteristics and functions compared to 2D monolayers of HERS cells in vitro. In vivo, HERS spheroids generated more mineralized tissue; when combined with dental papilla cells (DPCs), HERS spheroids contributed to dentin-like tissue formation. Moreover, the generation and expansion of HERS spheroids rely to some degree on the HIF-1 pathway. Conclusion: HERS spheroid generation is beneficial for functional HERS cell expansion and can provide a useful cell source for further tooth regeneration and mechanistic research. Notably, HIF-1 pathway plays a critical role in HERS spheroid formation and function.


Assuntos
Cultura Primária de Células/métodos , Endodontia Regenerativa/métodos , Esferoides Celulares/transplante , Raiz Dentária/crescimento & desenvolvimento , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Autorrenovação Celular , Dentina/metabolismo , Células Epiteliais/fisiologia , Células Epiteliais/transplante , Feminino , Fator 1 Induzível por Hipóxia/metabolismo , Modelos Animais , Odontogênese/fisiologia , Ratos , Regeneração , Esferoides Celulares/fisiologia , Células-Tronco/fisiologia
12.
Adv Biosyst ; 4(3): e1900254, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32293147

RESUMO

Islet transplantation has been demonstrated to be a promising therapy for type 1 diabetes mellitus. Although it is a minimally invasive operating procedure and provides easy access for graft monitoring, subcutaneous transplantation of the islet only has limited therapeutic outcomes, owing to the poor capacity of skin tissue to foster revascularization in a short period. Herein, 3D cell spheroids of clinically accessible umbilical cord blood mesenchymal stem cells and human umbilical vein endothelial cells are formed and employed for codelivery with ß cells subcutaneously. The 3D stem cell spheroids, which can secrete multiple proangiogenic and prosurvival growth factors, induce robust angiogenesis and prevent ß cell graft death, as indicated by the results of in vivo bioluminescent tracking and histological analysis. These experimental data highlight the efficacy of the 3D stem cell spheroids that are fabricated using translationally applicable cell types in promoting the survival and function of subcutaneously transplanted ß cells.


Assuntos
Sobrevivência Celular/fisiologia , Células Secretoras de Insulina , Neovascularização Fisiológica/fisiologia , Esferoides Celulares , Animais , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/transplante , Humanos , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/transplante , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Esferoides Celulares/citologia , Esferoides Celulares/transplante
13.
Methods Mol Biol ; 2140: 183-197, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32207113

RESUMO

Biomaterial-free three-dimensional (3D) bioprinting is a relatively new field within 3D bioprinting, where 3D tissues are created from the fusion of 3D multicellular spheroids, without requiring biomaterial. This is in contrast to traditional 3D bioprinting, which requires biomaterials to carry the cells to be bioprinted, such as a hydrogel or decellularized extracellular matrix. Here, we discuss principles of spheroid preparation for biomaterial-free 3D bioprinting of cardiac tissue. In addition, we discuss principles of using spheroids as building blocks in biomaterial-free 3D bioprinting, including spheroid dislodgement, spheroid transfer, and spheroid fusion. These principles are important considerations, to create the next generation of biomaterial-free spheroid-based 3D bioprinters.


Assuntos
Materiais Biocompatíveis , Bioimpressão/métodos , Miócitos Cardíacos , Impressão Tridimensional , Esferoides Celulares , Engenharia Tecidual/métodos , Alicerces Teciduais , Células Cultivadas , Citocinas/farmacologia , Fibroblastos , Células Endoteliais da Veia Umbilical Humana , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/transplante , Manejo de Espécimes , Esferoides Celulares/transplante
14.
Bull Exp Biol Med ; 168(4): 589-596, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32152851
15.
Cell Rep ; 30(3): 852-869.e4, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31968258

RESUMO

End-stage renal disease is a worldwide epidemic requiring renal replacement therapy. Harvesting tissue from failing kidneys and autotransplantation of tissue progenitors could theoretically delay the need for dialysis. Here we use healthy and end-stage human adult kidneys to robustly expand proliferative kidney epithelial cells and establish 3D kidney epithelial cultures termed "nephrospheres." Formation of nephrospheres reestablishes renal identity and function in primary cultures. Transplantation into NOD/SCID mice shows that nephrospheres restore self-organogenetic properties lost in monolayer cultures, allowing long-term engraftment as tubular structures, potentially adding nephron segments and demonstrating self-organization as critical to survival. Furthermore, long-term tubular engraftment of nephrospheres is functionally beneficial in murine models of chronic kidney disease. Remarkably, nephrospheres inhibit pro-fibrotic collagen production in cultured fibroblasts via paracrine modulation, while transplanted nephrospheres induce transcriptional signatures of proliferation and release from quiescence, suggesting re-activation of endogenous repair. These data support the use of human nephrospheres for renal cell therapy.


Assuntos
Rim/lesões , Rim/patologia , Esferoides Celulares/patologia , Cicatrização , Animais , Diferenciação Celular , Proliferação de Células , Doença Crônica , Modelos Animais de Doenças , Células Epiteliais/patologia , Fibrose , Humanos , Rim/fisiopatologia , Camundongos Endogâmicos NOD , Camundongos SCID , Insuficiência Renal Crônica/patologia , Esferoides Celulares/transplante
16.
Theranostics ; 9(22): 6587-6600, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31588237

RESUMO

It has been demonstrated that mesenchymal stem cells (MSCs) differentiated from human embryonic stem cells (hESCs), name EMSCs, can treat a variety of autoimmune and inflammatory diseases, with similar efficacies to those achieved with MSCs derived from somatic tissues such as bone marrow (BMSCs). The chance increases even higher for EMSCs, than somatic tissue derived MSCs​, to become a cell drug as the former can be produced in large scale from an unlimited hESC line with easier quality control and less biosafety concern. We have further demonstrated that both human ESCs and EMSCs, after aggregation to form spheroids, can tolerate hypoxic and ambient conditions (AC) for over 4 and 10 days, respectively, without loss of their viability and alteration of their functions. Based on these advantages, we decided to test whether EMSC spheroids, made in large quantity and delivered through a long-term distance at AC, can treat osteoarthritis spontaneously developed in rhesus macaques (M. mulatta) monkeys as well as the allogenic MSCs. Methods: Xenogeneic AC-transported EMSC spheroids or allogenic BMSCs were injected into the articular cavity of both knees of the monkeys at 3 animals per group. Another two macaques were injected the same way with saline as controls. Results: Both EMSCs and BMSCs groups showed significant amelioration indicated by the reduction of swelling joint size and amplification of keen flare angle post-treatment, compared to the control group. Examinations via X-ray and MRI also indicated the decrease of inflammation and osteophyma, and recovery of the synovium and cartilage in both treated groups. No sign of allergy or graft versus host disease was observed in the animals. Conclusion: Our results demonstrate that human EMSC spheroids can prevent the osteoarthtitis progression and ameliorate osteoarthritis in the rhesus macaques as well as allogenic BMSCs, and this study shall help advance the clinical application of EMSCs.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Osteoartrite/terapia , Animais , Células da Medula Óssea , Transplante de Medula Óssea , Diferenciação Celular , Condrócitos/citologia , Humanos , Articulação do Joelho/patologia , Macaca mulatta , Imageamento por Ressonância Magnética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Osteoartrite/diagnóstico por imagem , Esferoides Celulares/transplante
17.
Biomaterials ; 225: 119534, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31590118

RESUMO

3D culture of stem cells can improve therapeutic effects. However, there is limited research on how to deliver cultured stem cell spheroids to the desired target. Here, we developed lotus seedpod-inspired hydrogel (LoSH) containing microwells for culture and delivery of stem cell spheroids. Human adipose-derived stem cells (hADSCs) inside the square microwells (200 or 400 µm in width with various depths) spontaneously formed spheroids with high viability (94.08 ±â€¯1.56%), and fibronectins conjugated to the hydrogel successfully gripped the spheroids, similar to the funiculus gripping seeds in the lotus seedpod. The spheroids slightly bound to the LoSH surface at 37 °C were detached by the expansion of LoSH at lower temperature of 4 °C. After spheroid formation, LoSH was placed on the target substrate upside-down, expanded at 4 °C for 10 min, and removed from the target. As a result, the spheroids within the microwell were successfully transferred to the target substrate with high transfer efficiency (93.78 ±â€¯2.30%). A delivery of spheroids from LoSH to full-thickness murine skin wound with chimney model showed significant enhancement of the number of SMA-positive vessels at day 21 compared to the group received the same number of spheroids by injection. Together, our findings demonstrate LoSH as a one-step platform that can culture and deliver spheroids to a large target area, which will be useful for various biomedical applications.


Assuntos
Materiais Biomiméticos/farmacologia , Técnicas de Cultura de Células/métodos , Hidrogéis/farmacologia , Lotus/química , Sementes/química , Esferoides Celulares/transplante , Células-Tronco/citologia , Animais , Adesão Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Camundongos , Regeneração/efeitos dos fármacos , Pele/patologia , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Transplante de Células-Tronco , Células-Tronco/efeitos dos fármacos
18.
Nature ; 574(7776): 112-116, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31554966

RESUMO

Organogenesis is a complex and interconnected process that is orchestrated by multiple boundary tissue interactions1-7. However, it remains unclear how individual, neighbouring components coordinate to establish an integral multi-organ structure. Here we report the continuous patterning and dynamic morphogenesis of hepatic, biliary and pancreatic structures, invaginating from a three-dimensional culture of human pluripotent stem cells. The boundary interactions between anterior and posterior gut spheroids differentiated from human pluripotent stem cells enables retinoic acid-dependent emergence of hepato-biliary-pancreatic organ domains specified at the foregut-midgut boundary organoids in the absence of extrinsic factors. Whereas transplant-derived tissues are dominated by midgut derivatives, long-term-cultured microdissected hepato-biliary-pancreatic organoids develop into segregated multi-organ anlages, which then recapitulate early morphogenetic events including the invagination and branching of three different and interconnected organ structures, reminiscent of tissues derived from mouse explanted foregut-midgut culture. Mis-segregation of multi-organ domains caused by a genetic mutation in HES1 abolishes the biliary specification potential in culture, as seen in vivo8,9. In sum, we demonstrate that the experimental multi-organ integrated model can be established by the juxtapositioning of foregut and midgut tissues, and potentially serves as a tractable, manipulatable and easily accessible model for the study of complex human endoderm organogenesis.


Assuntos
Sistema Biliar/embriologia , Intestinos/embriologia , Fígado/embriologia , Modelos Biológicos , Morfogênese , Pâncreas/embriologia , Animais , Sistema Biliar/citologia , Biomarcadores/análise , Biomarcadores/metabolismo , Padronização Corporal , Endoderma/citologia , Endoderma/embriologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Intestinos/citologia , Fígado/citologia , Masculino , Camundongos , Organoides/citologia , Organoides/embriologia , Pâncreas/citologia , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Esferoides Celulares/transplante , Fatores de Transcrição HES-1/análise , Fatores de Transcrição HES-1/metabolismo
19.
Pediatr Surg Int ; 35(12): 1379-1388, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31552493

RESUMO

PURPOSE: Mesenchymal stem cell (MSC)-based cell therapies have emerged as a promising treatment option for various diseases. Due to the superior survival and higher differentiation efficiency, three-dimensional spheroid culture systems have been an important topic of MSC research. Stem cells from human exfoliated deciduous teeth (SHED) have been considered an ideal source of MSCs for regenerative medicine. Thus, in the present study, we introduce our newly developed method for fabricating SHED-based micro-hepatic tissues, and demonstrate the therapeutic effects of SHED-based micro-hepatic tissues in mouse disease models. METHODS: SHED-converted hepatocyte-like cells (SHED-HLCs) were used for fabricating spherical micro-hepatic tissues. The SHED-HLC-based spheroids were then transplanted both into the liver of mice with CCl4-induced chronic liver fibrosis and the kidney of factor VIII (F8)-knock-out mice. At 4 weeks after transplantation, the therapeutic efficacy was investigated. RESULTS: Intrahepatic transplantation of SHED-HLC-spheroids improved the liver dysfunction in association with anti-fibrosis effects in CCl4-treated mice. Transplanted SHED-converted cells were successfully engrafted in the recipient liver. Meanwhile, renal capsular transplantation of the SHED-HLC-spheroids significantly extended the bleeding time in F8-knock-out mice. CONCLUSIONS: These findings suggest that SHED-HLC-based micro-hepatic tissues might be a promising source for treating pediatric refractory diseases, including chronic liver fibrosis and hemophilia A.


Assuntos
Hemofilia A/terapia , Cirrose Hepática/terapia , Transplante de Células-Tronco Mesenquimais , Esferoides Celulares/transplante , Dente Decíduo , Transplante Heterólogo , Animais , Diferenciação Celular , Criança , Pré-Escolar , Doença Crônica , Modelos Animais de Doenças , Hepatócitos , Humanos , Masculino , Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos C57BL , Medicina Regenerativa/métodos
20.
Nat Commun ; 10(1): 3838, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31444335

RESUMO

Developing biomimetic nanoparticles without loss of the integrity of proteins remains a major challenge in cancer chemotherapy. Here, we develop a biocompatible tumor-cell-exocytosed exosome-biomimetic porous silicon nanoparticles (PSiNPs) as drug carrier for targeted cancer chemotherapy. Exosome-sheathed doxorubicin-loaded PSiNPs (DOX@E-PSiNPs), generated by exocytosis of the endocytosed DOX-loaded PSiNPs from tumor cells, exhibit enhanced tumor accumulation, extravasation from blood vessels and penetration into deep tumor parenchyma following intravenous administration. In addition, DOX@E-PSiNPs, regardless of their origin, possess significant cellular uptake and cytotoxicity in both bulk cancer cells and cancer stem cells (CSCs). These properties endow DOX@E-PSiNPs with great in vivo enrichment in total tumor cells and side population cells with features of CSCs, resulting in anticancer activity and CSCs reduction in subcutaneous, orthotopic and metastatic tumor models. These results provide a proof-of-concept for the use of exosome-biomimetic nanoparticles exocytosed from tumor cells as a promising drug carrier for efficient cancer chemotherapy.


Assuntos
Doxorrubicina/administração & dosagem , Portadores de Fármacos/química , Composição de Medicamentos/métodos , Exossomos/química , Neoplasias/tratamento farmacológico , Animais , Linhagem Celular Tumoral/citologia , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/transplante , Modelos Animais de Doenças , Exocitose , Feminino , Humanos , Masculino , Camundongos , Nanopartículas/química , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Porosidade , Estudo de Prova de Conceito , Silício/química , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Esferoides Celulares/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA