Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
FASEB J ; 36(11): e22613, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36250916

RESUMO

Intestinal failure (IF) occurs when intestinal surface area or function is not sufficient to support digestion and nutrient absorption. Human intestinal organoid (HIO)-derived tissue-engineered intestine is a potential cure for IF. Research to date has demonstrated successful HIO transplantation (tHIO) into mice with significant in vivo maturation. An area lacking in the literature is exploration of murine host sex as a biological variable (SABV) in tHIO function. In this study, we investigate murine host SABV in tHIO epithelial barrier function and muscle contractility. HIOs were generated in vitro and transplanted into nonobese diabetic, severe combined immunodeficiency gamma chain deficient male and female mice. tHIOs were harvested after 8-12 weeks in vivo. Reverse transcriptase polymerase chain reaction and immunohistochemistry were conducted to compare tight junctions and contractility-related markers in tHIOs. An Ussing chamber and contractility apparatus were used to evaluate tHIO epithelial barrier and muscle contractile function, respectively. The expression and morphology of tight junction and contractility-related markers from tHIOs in male and female murine hosts is not significantly different. Epithelial barrier function as measured by transepithelial resistance, short circuit current, and fluorescein isothiocyanate-dextran permeability is no different in tHIOs from male and female hosts, although these results may be limited by HIO epithelial immaturity and a short flux time. Muscle contractility as measured by total contractile activity, amplitude, frequency, and tension is not significantly different in tHIOs from male and female hosts. The data suggest that murine host sex may not be a significant biological variable influencing tHIO function, specifically epithelial barrier maintenance and muscle contractility, though limitations exist in our model.


Assuntos
Dextranos , Organoides , Animais , Dextranos/metabolismo , Feminino , Humanos , Mucosa Intestinal/metabolismo , Intestinos , Masculino , Camundongos , Músculos/metabolismo , Organoides/metabolismo , Permeabilidade , Junções Íntimas/metabolismo
2.
Dig Dis Sci ; 67(12): 5511-5521, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35334015

RESUMO

BACKGROUND: Human intestinal organoids (HIOs), when transplanted into immunocompromised mice (tHIOs), demonstrate significant growth and maturation. While both male and female mice are reported to be viable hosts for these experiments, a direct comparison of sex-related differences in tHIO structure and development has not been performed. AIMS: We sought to identify host sex-related differences in tHIO engraftment, morphology, and epithelial and mesenchymal development. METHODS: HIOs were generated in vitro and transplanted beneath the kidney capsule of NSG male and female mice. tHIOs were harvested at 8-9 weeks. Anthropometric measurements were captured. tHIOs were divided in half and histology or RT-qPCR performed. Morphology was evaluated and epithelial architecture graded on a scale of 1 (absence of crypts/villi) to 4 (elongated crypt-villus axis). RT-qPCR and immunofluorescence microscopy were performed for epithelial and mesenchymal differentiation markers. RESULTS: Host survival and tHIO engraftment were equivalent in male and female hosts. tHIO weight and length were also equivalent between groups. The number of lumens per tHIOs from male and female hosts was similar, but the mean lumen circumference was larger for tHIOs from male hosts. tHIOs from male hosts were more likely to demonstrate higher grades of epithelial development. However, both groups showed similar differentiation into secretory and absorptive epithelial lineages. Markers for intestinal identity, mesenchymal development, and brush border enzymes were also expressed similarly between groups. CONCLUSIONS: While male host sex was associated with larger tHIO lumen size and mucosal maturation, tHIOs from both groups had similar engraftment, growth, and epithelial and mesenchymal cytodifferentiation.


Assuntos
Organoides , Transplantes , Humanos , Masculino , Feminino , Camundongos , Animais , Organoides/patologia , Organoides/transplante , Intestinos , Mucosa Intestinal , Microvilosidades
3.
Cytotherapy ; 23(5): 381-389, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33840629

RESUMO

The field of regenerative medicine is developing technologies that, in the near future, will offer alternative approaches to either cure diseases affecting the gastrointestinal tract or slow their progression by leveraging the intrinsic ability of our tissues and organs to repair after damage. This article will succinctly illustrate the three technologies that are closer to clinical translation-namely, human intestinal organoids, sphincter bioengineering and decellularization, whereby the cellular compartment of a given segment of the digestive tract is removed to obtain a scaffold consisting of the extracellular matrix. The latter will be used as a template for the regeneration of a functional organ, whereby the newly generated cellular compartment will be obtained from the patient's own cells. Although clinical application of this technology is approaching, product development challenges are being tackled to warrant safety and efficacy.


Assuntos
Engenharia Tecidual , Alicerces Teciduais , Bioengenharia , Matriz Extracelular , Trato Gastrointestinal , Humanos , Medicina Regenerativa
4.
J Surg Res ; 259: 500-508, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33168233

RESUMO

BACKGROUND: Short bowel syndrome is a potentially fatal condition with inadequate management options. Tissue-engineered small intestine (TESI) is a promising solution, but confirmation of TESI function will be crucial before human application. We sought to define intestinal epithelial barrier function in human intestinal organoid (HIO)-derived TESI. MATERIALS AND METHODS: HIOs were generated in vitro from human embryonic stem cells. After 1 mo, HIOs were collected for analysis or transplanted into the kidney capsule of immunocompromised mice. Transplanted HIOs (tHIOs) were harvested for analysis at 4 or 8 wk. Reverse transcription quantitative polymerase chain reaction and immunofluorescent staining were performed for tight junction components: claudin 3 (CLDN3), claudin 15 (CLDN15), occludin (OCLN), and zonula occludens-1, or tight junction protein-1 (TJP1/ZO-1). RESULTS: Four-week-old tHIOs demonstrated significantly (P < 0.05) higher levels of CLDN15 (6x), OCLN (4x), and TJP1/ZO-1 (3x) normalized to GAPDH than in vitro HIOs. Eight-week-old tHIOs demonstrated significantly (P < 0.05) higher expression levels of CLDN3 (26x), CLDN15 (29x), OCLN (4x), and TJP1/ZO-1 (5x) than in vitro HIOs. There was no significant difference in expression of these tight junction components between 4- and 8-week-old tHIOs. Immunofluorescent staining revealed the presence of claudin 3, claudin 15, occludin, and zonula occludens-1 in both in vitro HIOs and tHIOs; however, the morphology appeared more mature in tHIOs. CONCLUSIONS: In vitro HIOs have lower levels of tight junction mRNA, and tight junction proteins appear morphologically immature. Transplantation facilitates maturation of the HIOs and enhances select tight junction gene expression.


Assuntos
Intestinos/citologia , Organoides/transplante , Síndrome do Intestino Curto/cirurgia , Proteínas de Junções Íntimas/metabolismo , Engenharia Tecidual , Animais , Técnicas de Cultura de Células/métodos , Linhagem Celular , Regulação da Expressão Gênica , Células-Tronco Embrionárias Humanas , Humanos , Masculino , Camundongos , Modelos Animais , Junções Íntimas/metabolismo
5.
Calcif Tissue Int ; 100(3): 298-310, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27913821

RESUMO

Theobromine (THB) is one of the major xanthine-like alkaloids found in cacao plant and a variety of other foodstuffs such as tea leaves, guarana and cola nuts. Historically, THB and its derivatives have been utilized to treat cardiac and circulatory disorders, drug-induced nephrotoxicity, proteinuria and as an immune-modulator. Our previous work demonstrated that THB has the capacity to improve the formation of hydroxyl-apatite during tooth development, suggesting that it may also enhance skeletal development. With its excellent safety profile and resistance to pharmacokinetic elimination, we reasoned that it might be an excellent natural osteoanabolic supplement during pregnancy, lactation and early postnatal growth. To determine whether THB had an effect on human osteoprogenitors, we subjected primary human bone marrow mesenchymal stem cells (hMSCs) to osteogenic assays after exposure to THB in vitro and observed that THB exposure increased the rate of osteogenesis and mineralization by hMSCs. Moreover, THB exposure resulted in a list of upregulated mRNA transcripts that best matched an osteogenic tissue expression signature as compared to other tissue expression signatures archived in several databases. To determine whether oral administration of THB resulted in improved skeletal growth, we provided pregnant rats with chow supplemented with THB during pregnancy and lactation. After weaning, offspring received THB continuously until postnatal day 50 (approximately 10 mg kg-1 day-1). Administration of THB resulted in neonates with larger bones, and 50-day-old offspring accumulated greater body mass, longer and thicker femora and superior tibial trabecular parameters. The accelerated growth did not adversely affect the strength and resilience of the bones. These results indicate that THB increases the osteogenic potential of bone marrow osteoprogenitors, and dietary supplementation of a safe dose of THB to expectant mothers and during the postnatal period could accelerate skeletal development in their offspring.


Assuntos
Desenvolvimento Ósseo/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Teobromina/farmacologia , Animais , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo , Ratos , Regulação para Cima/efeitos dos fármacos
6.
Front Bioeng Biotechnol ; 10: 1015628, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36263358

RESUMO

Regenerative medicine (RM) is changing how we think and practice transplant medicine. In regenerative medicine, the aim is to develop and employ methods to regenerate, restore or replace damaged/diseased tissues or organs. Regenerative medicine investigates using tools such as novel technologies or techniques, extracellular vesicles, cell-based therapies, and tissue-engineered constructs to design effective patient-specific treatments. This review illustrates current advancements in regenerative medicine that may pertain to transplant medicine. We highlight progress made and various tools designed and employed specifically for each tissue or organ, such as the kidney, heart, liver, lung, vasculature, gastrointestinal tract, and pancreas. By combing both fields of transplant and regenerative medicine, we can harbor a successful collaboration that would be beneficial and efficacious for the repair and design of de novo engineered whole organs for transplantations.

7.
Front Bioeng Biotechnol ; 9: 764703, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34796168

RESUMO

Novel bone regeneration strategies often show promise in rodent models yet are unable to successfully translate to clinical therapy. Sheep, goats, and dogs are used as translational models in preparation for human clinical trials. While human MSCs (hMSCs) undergo osteogenesis in response to well-defined protocols, canine MSCs (cMSCs) are more incompletely characterized. Prior work suggests that cMSCs require additional agonists such as IGF-1, NELL-1, or BMP-2 to undergo robust osteogenic differentiation in vitro. When compared directly to hMSCs, cMSCs perform poorly in vivo. Thus, from both mechanistic and clinical perspectives, cMSC and hMSC-mediated bone regeneration may differ. The objectives of this study were twofold. The first was to determine if previous in vitro findings regarding cMSC osteogenesis were substantiated in vivo using an established murine calvarial defect model. The second was to assess in vitro ALP activity and endogenous BMP-2 gene expression in both canine and human MSCs. Calvarial defects (4 mm) were treated with cMSCs, sub-therapeutic BMP-2, or the combination of cMSCs and sub-therapeutic BMP-2. At 28 days, while there was increased healing in defects treated with cMSCs, defects treated with cMSCs and BMP-2 exhibited the greatest degree of bone healing as determined by quantitative µCT and histology. Using species-specific qPCR, cMSCs were not detected in relevant numbers 10 days after implantation, suggesting that bone healing was mediated by anabolic cMSC or ECM-driven cues and not via engraftment of cMSCs. In support of this finding, defects treated with cMSC + BMP-2 exhibited robust deposition of Collagens I, III, and VI using immunofluorescence. Importantly, cMSCs exhibited minimal ALP activity unless cultured in the presence of BMP-2 and did not express endogenous canine BMP-2 under any condition. In contrast, human MSCs exhibited robust ALP activity in all conditions and expressed human BMP-2 when cultured in control and osteoinduction media. This is the first in vivo study in support of previous in vitro findings regarding cMSC osteogenesis, namely that cMSCs require additional agonists to initiate robust osteogenesis. These findings are highly relevant to translational cell-based bone healing studies and represent an important finding for the field of canine MSC-mediated bone regeneration.

8.
J Med Imaging (Bellingham) ; 8(1): 014503, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33542945

RESUMO

Purpose: Mesenchymal stem cells (MSCs) have demonstrated clinically relevant therapeutic effects for treatment of trauma and chronic diseases. The proliferative potential, immunomodulatory characteristics, and multipotentiality of MSCs in monolayer culture is reflected by their morphological phenotype. Standard techniques to evaluate culture viability are subjective, destructive, or time-consuming. We present an image analysis approach to objectively determine morphological phenotype of MSCs for prediction of culture efficacy. Approach: The algorithm was trained using phase-contrast micrographs acquired during the early and mid-logarithmic stages of MSC expansion. Cell regions are localized using edge detection, thresholding, and morphological operations, followed by cell marker identification using H-minima transform within each region to differentiate individual cells from cell clusters. Clusters are segmented using marker-controlled watershed to obtain single cells. Morphometric and textural features are extracted to classify cells based on phenotype using machine learning. Results: Algorithm performance was validated using an independent test dataset of 186 MSCs in 36 culture images. Results show 88% sensitivity and 86% precision for overall cell detection and a mean Sorensen-Dice coefficient of 0.849 ± 0.106 for segmentation per image. The algorithm exhibited an area under the curve of 0.816 ( CI 95 = 0.769 to 0.886) and 0.787 ( CI 95 = 0.716 to 0.851) for classifying MSCs according to their phenotype at early and mid-logarithmic expansion, respectively. Conclusions: The proposed method shows potential to segment and classify low and moderately dense MSCs based on phenotype with high accuracy and robustness. It enables quantifiable and consistent morphology-based quality assessment for various culture protocols to facilitate cytotherapy development.

9.
Methods Cell Biol ; 156: 15-43, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32222217

RESUMO

Bone is a composite material consisting primarily of cells, extracellular matrices, accessory proteins and the complex calcium phosphate salt hydroxyapatite. Collectively, the extracellular network of proteins and accessory molecules that provide the organic component of bone tissue is referred to as the osteogenic extracellular matrix (OECM). OECM provides tensile strength and increases the durability of bone, but the OECM also serves as an attachment site and regulatory substrate for cells and a repository for growth factors and cytokines. Increasingly, purified OECM generated by osteogenic cells in culture has attracted interest because it has the capacity to improve the growth and viability of attached cells, enhances the osteogenic program in vitro and in vivo, and shows great promise as a therapeutic tool for orthopedic tissue engineering. This chapter will describe fundamental protocols for the selection and culture of osteogenic cells and conditions for their osteogenic differentiation, and the synthesis, purification and characterization of OECM. Some examples of immobilization to surfaces for the purpose of two- and three-dimensional culture will also be described.


Assuntos
Matriz Extracelular/metabolismo , Osteogênese , Engenharia Tecidual/métodos , Fosfatase Alcalina/metabolismo , Animais , Biomarcadores/metabolismo , Calcificação Fisiológica , Adesão Celular , Células Cultivadas , Matriz Extracelular/ultraestrutura , Humanos , Células-Tronco Mesenquimais/citologia , Suínos
10.
Adv Healthc Mater ; 9(15): e1901580, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32147960

RESUMO

Additive manufacturing is a promising method for producing customized 3D bioactive constructs for regenerative medicine. Here, 3D printed highly osteogenic scaffolds using nanoengineered ionic-covalent entanglement ink (NICE) for bone tissue engineering are reported. This NICE ink consists of ionic-covalent entanglement reinforced with Laponite, a 2D nanosilicate (nSi) clay, allowing for the printing of anatomic-sized constructs with high accuracy. The 3D printed structure is able to maintain high structural stability in physiological conditions without any significant swelling or deswelling. The presence of nSi imparts osteoinductive characteristics to the NICE scaffolds, which is further augmented by depositing pluripotent stem cell-derived extracellular matrix (ECM) on the scaffolds. This is achieved by stimulating human induced pluripotent stem cell-derived mesenchymal stem cells (iP-hMSCs) with 2-chloro-5-nitrobenzanilide, a PPARγ inhibitor that enhances Wnt pathway, resulting in the deposition of an ECM characterized by high levels of collagens VI and XII found in anabolic bone. The osteoinductive characteristics of these bioconditioned NICE (bNICE) scaffolds is demonstrated through osteogenic differentiation of bone marrow derived human mesenchymal stem cells. A significant increase in the expression of osteogenic gene markers as well as mineralized ECM are observed on bioconditioned NICE (bNICE) scaffolds compared to bare scaffolds (NICE). The bioconditioned 3D printed scaffolds provide a unique strategy to design personalized bone grafts for in situ bone regeneration.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Mesenquimais , Diferenciação Celular , Humanos , Osteogênese , Impressão Tridimensional , Engenharia Tecidual , Alicerces Teciduais
11.
Nat Commun ; 11(1): 3025, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32541821

RESUMO

Approximately 10% of fractures will not heal without intervention. Current treatments can be marginally effective, costly, and some have adverse effects. A safe and manufacturable mimic of anabolic bone is the primary goal of bone engineering, but achieving this is challenging. Mesenchymal stem cells (MSCs), are excellent candidates for engineering bone, but lack reproducibility due to donor source and culture methodology. The need for a bioactive attachment substrate also hinders progress. Herein, we describe a highly osteogenic MSC line generated from induced pluripotent stem cells that generates high yields of an osteogenic cell-matrix (ihOCM) in vitro. In mice, the intrinsic osteogenic activity of ihOCM surpasses bone morphogenic protein 2 (BMP2) driving healing of calvarial defects in 4 weeks by a mechanism mediated in part by collagen VI and XII. We propose that ihOCM may represent an effective replacement for autograft and BMP products used commonly in bone tissue engineering.


Assuntos
Osteogênese , Células-Tronco Pluripotentes/citologia , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Proliferação de Células , Células Cultivadas , Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Colágeno Tipo XII/genética , Colágeno Tipo XII/metabolismo , Anormalidades Craniofaciais/fisiopatologia , Anormalidades Craniofaciais/terapia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante , Engenharia Tecidual
12.
Cell Death Dis ; 9(12): 1161, 2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30478297

RESUMO

Malignant bone disease (MBD) occurs when tumors establish in bone, causing catastrophic tissue damage as a result of accelerated bone destruction and inhibition of repair. The resultant so-called osteolytic lesions (OL) take the form of tumor-filled cavities in bone that cause pain, fractures, and associated morbidity. Furthermore, the OL microenvironment can support survival of tumor cells and resistance to chemotherapy. Therefore, a deeper understanding of OL formation and MBD progression is imperative for the development of future therapeutic strategies. Herein, we describe a novel in vitro platform to study bone-tumor interactions based on three-dimensional co-culture of osteogenically enhanced human mesenchymal stem cells (OEhMSCs) in a rotating wall vessel bioreactor (RWV) while attached to micro-carrier beads coated with extracellular matrix (ECM) composed of factors found in anabolic bone tissue. Osteoinhibition was recapitulated in this model by co-culturing the OEhMSCs with a bone-tumor cell line (MOSJ-Dkk1) that secretes the canonical Wnt (cWnt) inhibitor Dkk-1, a tumor-borne osteoinhibitory factor widely associated with several forms of MBD, or intact tumor fragments from Dkk-1 positive patient-derived xenografts (PDX). Using the model, we observed that depending on the conditions of growth, tumor cells can biochemically inhibit osteogenesis by disrupting cWnt activity in OEhMSCs, while simultaneously co-engrafting with OEhMSCs, displacing them from the niche, perturbing their activity, and promoting cell death. In the absence of detectable co-engraftment with OEhMSCs, Dkk-1 positive PDX fragments had the capacity to enhance OEhMSC proliferation while inhibiting their osteogenic differentiation. The model described has the capacity to provide new and quantifiable insights into the multiple pathological mechanisms of MBD that are not readily measured using monolayer culture or animal models.


Assuntos
Doenças Ósseas/genética , Neoplasias Ósseas/genética , Células-Tronco Mesenquimais/metabolismo , Osteogênese/genética , Animais , Reatores Biológicos , Doenças Ósseas/patologia , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Técnicas de Cultura de Células , Diferenciação Celular/genética , Proliferação de Células/genética , Técnicas de Cocultura , Matriz Extracelular/genética , Matriz Extracelular/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Células-Tronco Mesenquimais/patologia , Osteólise/genética , Osteólise/patologia , Microambiente Tumoral/genética , Via de Sinalização Wnt/genética
13.
Spine J ; 17(3): 418-430, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27765715

RESUMO

BACKGROUND CONTEXT: Spine pain and the disability associated with it are epidemic in the United States. According to the National Center for Health Statistics, more than 650,000 spinal fusion surgeries are performed annually in the United States, and yet there is a failure rate of 15%-40% when standard methods employing current commercial bone substitutes are used. Autologous bone graft is the gold standard in terms of fusion success, but the morbidity associated with the procedure and the limitations in the availability of sufficient material have limited its use in the majority of cases. A freely available and immunologically compatible bone mimetic with the properties of live tissue is likely to substantially improve the outcome of spine fusion procedures without the disadvantages of autologous bone graft. PURPOSE: This study aimed to compare a live human bone tissue analog with autologous bone grafting in an immunocompromised rat model of posterolateral fusion. DESIGN/SETTING: This is an in vitro and in vivo preclinical study of a novel human stem cell-derived construct for efficacy in posterolateral lumbar spine fusion. METHODS: Osteogenically enhanced human mesenchymal stem cells (OEhMSCs) were generated by exposure to conditions that activate the early stages of osteogenesis. Immunologic characteristics of OEhMSCs were evaluated in vitro. The secreted extracellular matrix from OEhMSCs was deposited on a clinical-grade gelatin sponge, resulting in bioconditioned gelatin sponge (BGS). Bioconditioned gelatin sponge was used alone, with live OEhMSCs (BGS+OEhMSCs), or with whole human bone marrow (BGS+hBM). Efficacy for spine fusion was determined by an institutionally approved animal model using 53 nude rats. RESULTS: Bioconditioned gelatin sponge with live OEhMSCs did not cause cytotoxicity when incubated with immunologically mismatched lymphocytes, and OEhMSCs inhibited lymphocyte expansion in mixed lymphocyte assays. Bioconditioned gelatin sponge with live OEhMSC and BGS+hBM constructs induced profound bone growth at fusion sites in vivo, with a comparable rate of fusion with syngeneic bone graft (negative [0 of 10], BGS alone [0 of 10], bone graft [7 of 10], BGS+OEhMSC [10 of 15], and BGS+hBM [8 of 8]). CONCLUSIONS: Collectively, these studies demonstrate that BGS+OEhMSC constructs possess low immunogenicity and drive vertebral fusion with efficiency matching syngeneic bone graft in rodents. We also demonstrate that BGS serves as a promising scaffold for spine fusion when combined with hBM.


Assuntos
Células-Tronco Adultas , Aloenxertos , Substitutos Ósseos , Transplante Ósseo/métodos , Vértebras Lombares/cirurgia , Células-Tronco Mesenquimais , Fusão Vertebral/métodos , Adulto , Animais , Feminino , Gelatina , Humanos , Vértebras Lombares/fisiologia , Modelos Animais , Osteogênese , Ratos Nus , Transplante Autólogo , Transplante Homólogo
14.
J Bone Miner Res ; 30(1): 83-94, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25130615

RESUMO

Although bone has remarkable regenerative capacity, about 10% of long bone fractures and 25% to 40% of vertebral fusion procedures fail to heal. In such instances, a scaffold is employed to bridge the lesion and accommodate osteoprogenitors. Although synthetic bone scaffolds mimic some of the characteristics of bone matrix, their effectiveness can vary because of biological incompatibility. Herein, we demonstrate that a composite prepared with osteogenically enhanced mesenchymal stem cells (OEhMSCs) and their extracellular matrix (ECM) has an unprecedented capacity for the repair of critical-sized defects of murine femora. Furthermore, OEhMSCs do not cause lymphocyte activation, and ECM/OEhMSC composites retain their in vivo efficacy after cryopreservation. Finally, we show that attachment to the ECM by OEhMSCs stimulates the production of osteogenic and angiogenic factors. These data demonstrate that composites of OEhMSCs and their ECM could be utilized in the place of autologous bone graft for complex orthopedic reconstructions.


Assuntos
Regeneração Óssea , Criopreservação , Proteínas da Matriz Extracelular/biossíntese , Matriz Extracelular/química , Células-Tronco Mesenquimais/metabolismo , Alicerces Teciduais/química , Animais , Células Cultivadas , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA