Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Cell Biochem ; 118(11): 3627-3634, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28621436

RESUMO

Cell and gene therapy approaches are safer when they possess a system that enables the therapy to be rapidly halted. Human mesenchymal stem cells were transduced with an adenoviral vector containing the cDNA for bone morphogenetic protein 2 (AdBMP2) to induce bone formation. To make this method safer, a system to quickly kill these virally transduced cells was designed and evaluated. Cells were encapsulated inside poly(ethylene glycol) diacrylate (PEG-Da) hydrogels that are able to shield the cells from immunological destruction. The system involves an inducible caspase-9 (iCasp9) activated using a specific chemical inducer of dimerization (CID). Delivering AdBMP2-transduced human mesenchymal stem cells encapsulated in PEG-Da hydrogel promoted ectopic ossification in vivo, and the iCasp9 system allowed direct control of the timing of apoptosis of the injected cells. The iCasp9-CID system enhances the safety of delivering AdBMP2-transduced cells, making it a more compelling therapeutic for bone repair and spine fusion. J. Cell. Biochem. 118: 3627-3634, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Adenoviridae , Proteína Morfogenética Óssea 2 , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Genética/métodos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Fusão Vertebral/métodos , Transdução Genética , Animais , Proteína Morfogenética Óssea 2/biossíntese , Proteína Morfogenética Óssea 2/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
2.
Bio Protoc ; 14(3): e4931, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38379831

RESUMO

Recombinant adeno-associated viruses (rAAVs) are valuable viral vectors for in vivo gene transfer, also having significant ex vivo therapeutic potential. Continued efforts have focused on various gene therapy applications, capsid engineering, and scalable manufacturing processes. Adherent cells are commonly used for virus production in most basic science laboratories because of their efficiency and cost. Although suspension cells are easier to handle and scale up compared to adherent cells, their use in virus production is hampered by poor transfection efficiency. In this protocol, we developed a simple scalable AAV production protocol using serum-free-media-adapted HEK293T suspension cells and VirusGEN transfection reagent. The established protocol allows AAV production from transfection to quality analysis of purified AAV within two weeks. Typical vector yields for the described suspension system followed by iodixanol purification range from a total of 1 × 1013 to 1.5 × 1013 vg (vector genome) using 90 mL of cell suspension vs. 1 × 1013 to 2 × 1013 vg using a regular adherent cell protocol (10 × 15 cm dishes). Key features • Adeno-associated virus (AAV) production using serum-free-media-adapted HEK293T suspension cells. • Efficient transfection with VirusGEN. • High AAV yield from small-volume cell culture. Graphical overview.

3.
J Cell Biochem ; 112(10): 2748-58, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21678472

RESUMO

Heterotopic ossification (HO), or bone formation in soft tissues, is often the result of traumatic injury. Much evidence has linked the release of BMPs (bone morphogenetic proteins) upon injury to this process. HO was once thought to be a rare occurrence, but recent statistics from the military suggest that as many as 60% of traumatic injuries, resulting from bomb blasts, have associated HO. In this study, we attempt to define the role of peripheral nerves in this process. Since BMP2 has been shown previously to induce release of the neuroinflammatory molecules, substance P (SP) and calcitonin gene related peptide (CGRP), from peripheral, sensory neurons, we examined this process in vivo. SP and CGRP are rapidly expressed upon delivery of BMP2 and remain elevated throughout bone formation. In animals lacking functional sensory neurons (TRPV1(-/-) ), BMP2-mediated increases in SP and CGRP were suppressed as compared to the normal animals, and HO was dramatically inhibited in these deficient mice, suggesting that neuroinflammation plays a functional role. Mast cells, known to be recruited by SP and CGRP, were elevated after BMP2 induction. These mast cells were localized to the nerve structures and underwent degranulation. When degranulation was inhibited using cromolyn, HO was again reduced significantly. Immunohistochemical analysis revealed nerves expressing the stem cell markers nanog and Klf4, as well as the osteoblast marker osterix, after BMP2 induction, in mice treated with cromolyn. The data collectively suggest that BMP2 can act directly on sensory neurons to induce neurogenic inflammation, resulting in nerve remodeling and the migration/release of osteogenic and other stem cells from the nerve. Further, blocking this process significantly reduces HO, suggesting that the stem cell population contributes to bone formation.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Inflamação Neurogênica/complicações , Inflamação Neurogênica/fisiopatologia , Ossificação Heterotópica/etiologia , Ossificação Heterotópica/metabolismo , Células Receptoras Sensoriais/patologia , Animais , Proteína Morfogenética Óssea 2/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Linhagem Celular , Cromolina Sódica/farmacologia , Imuno-Histoquímica , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos Endogâmicos C57BL , Ossificação Heterotópica/genética , Células Receptoras Sensoriais/imunologia , Substância P/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Microtomografia por Raio-X
4.
J Cell Biochem ; 112(6): 1563-71, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21344484

RESUMO

More than a decade has passed since the first experiments using adenovirus-transduced cells expressing bone morphogenetic protein 2 were performed for the synthesis of bone. Since this time, the field of bone gene therapy has tackled many issues surrounding safety and efficacy of this type of strategy. We present studies examining the parameters of the timing of bone healing, and remodeling when heterotopic ossification (HO) is used for bone fracture repair using an adenovirus gene therapy approach. We use a rat fibula defect, which surprisingly does not heal even when a simple fracture is introduced. In this model, the bone quickly resorbs most likely due to the non-weight bearing nature of this bone in rodents. Using our gene therapy system robust HO can be introduced at the targeted location of the defect resulting in bone repair. The HO and resultant bone healing appeared to be dose dependent, based on the number of AdBMP2-transduced cells delivered. Interestingly, the HO undergoes substantial remodeling, and assumes the size and shape of the missing segment of bone. However, in some instances we observed some additional bone associated with the repair, signifying that perhaps the forces on the newly forming bone are inadequate to dictate shape. In all cases, the HO appeared to fuse into the adjacent long bone. The data collectively indicates that the use of BMP2 gene therapy strategies may vary depending on the location and nature of the defect. Therefore, additional parameters should be considered when implementing such strategies.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Fíbula/anormalidades , Terapia Genética/métodos , Adenoviridae/genética , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Osso e Ossos/anormalidades , Linhagem Celular , Humanos , Camundongos , Osteogênese/fisiologia , Ratos , Cicatrização/fisiologia
5.
Stem Cells Transl Med ; 10(4): 623-635, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33245845

RESUMO

Bone morphogenetic protein 2 (BMP2)-induced heterotopic bone formation (HBF) starts synchronously from zero upon BMP2 induction, which is advantageous for lineage tracking. The studies reported here in GLAST-CreErt2 :tdTomato red (TR)floxSTOPflox mice during BMP2-induced HBF show 78.8 ± 11.6% of chondrocytes and 86.5 ± 1.9% of osteoblasts are TR+ after approximately 1 week. Clustering after single-cell RNAseq resulted in nine cell types, and analysis revealed one as a highly replicating stem-like cell (RSC). Pseudotiming suggested that the RSC transitions to a mesenchymal stem-like cell that simultaneously expresses multiple osteoblast and chondrocyte transcripts (chondro-osseous progenitor [COP]). RSCs and COPs were isolated using flow cytometry for unique surface markers. Isolated RSCs (GLAST-TR+ Hmmr+ Cd200- ) and COPs (GLAST-TR+ Cd200+ Hmmr- ) were injected into the muscle of mice undergoing HBF. Approximately 9% of the cells in heterotopic bone (HB) in mice receiving RSCs were GLAST-TR+ , compared with less than 0.5% of the cells in mice receiving COPs, suggesting that RSCs are many times more potent than COPs. Analysis of donor-derived TR+ RSCs isolated from the engrafted HB showed approximately 50% were COPs and 45% were other cells, presumably mature bone cells, confirming the early nature of the RSCs. We next isolated RSCs from these mice (approximately 300) and injected them into a second animal, with similar findings upon analysis of HBF. Unlike other methodology, single cell RNAseq has the ability to detect rare cell populations such as RSCs. The fact that RSCs can be injected into mice and differentiate suggests their potential utility for tissue regeneration.


Assuntos
Proteína Morfogenética Óssea 2 , Ossificação Heterotópica , Células-Tronco , Animais , Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular , Células-Tronco Mesenquimais , Camundongos , Osteoblastos , Células-Tronco/citologia
6.
Crit Rev Eukaryot Gene Expr ; 20(4): 313-24, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21395504

RESUMO

Heterotopic ossification, defined as the formation of bone in abnormal anatomic locations, can be clinically insignificant or devastating and debilitating, depending on the site and duration of new bone formation. There are many causes of heterotopic ossification (HO), including soft tissue trauma, central nervous system injury, vasculopathies, arthropathies, and inheritance. One of the least understood components of HO is the interaction of the peripheral nervous system with the induction of this process. Recent work has shown that, upon traumatic injury, a cascade of events termed neurogenic inflammation is initiated, which involves the release of neuropeptides, such as substance P and calcitonin gene related peptide. Release of these peptides ultimately leads to the recruitment of activated platelets, mast cells, and neutrophils to the injury site. These cells appear to be involved with both remodeling of the nerve, as well as potentially recruiting additional cells from the bone marrow to the injury site. Further, sensory neurons stimulated at the injury site relay local information to the brain, which can then redirect neuroendocrine signaling in the hypothalamus towards repair of the injured site. While numerous studies have highlighted the important role of nerve-derived signals, both central and peripheral, in the regulation of normal bone remodeling of the skeleton,1 this review focuses on the role of the local, peripheral nerves in the formation of heterotopic bone. We concentrate on the manner in which local changes in bone morphogenetic protein (BMP) expression contribute to a cascade of events within the peripheral nerves, both sensory and sympathetic, in the immediate area of HO formation.


Assuntos
Ossificação Heterotópica/etiologia , Ossificação Heterotópica/fisiopatologia , Sistema Nervoso Periférico/fisiopatologia , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Humanos , Inflamação/fisiopatologia , Camundongos , Modelos Neurológicos , Neuropeptídeos/fisiologia , Osteoblastos/fisiologia , Transdução de Sinais , Canais de Cátion TRPV/fisiologia
7.
J Cell Biol ; 163(5): 1133-43, 2003 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-14662751

RESUMO

Once escaped from the quiescence niche, precursor cells interact with stromal components that support their survival, proliferation, and differentiation. We examined interplays between human myogenic precursor cells (mpc) and monocyte/macrophages (MP), the main stromal cell type observed at site of muscle regeneration. mpc selectively and specifically attracted monocytes in vitro after their release from quiescence, chemotaxis declining with differentiation. A DNA macroarray-based strategy identified five chemotactic factors accounting for 77% of chemotaxis: MP-derived chemokine, monocyte chemoattractant protein-1, fractalkine, VEGF, and the urokinase system. MP showed lower constitutive chemotactic activity than mpc, but attracted monocytes much strongly than mpc upon cross-stimulation, suggesting mpc-induced and predominantly MP-supported amplification of monocyte recruitment. Determination of [3H]thymidine incorporation, oligosomal DNA levels and annexin-V binding showed that MP stimulate mpc proliferation by soluble factors, and rescue mpc from apoptosis by direct contacts. We conclude that once activated, mpc, which are located close by capillaries, initiate monocyte recruitment and interplay with MP to amplify chemotaxis and enhance muscle growth.


Assuntos
Apoptose , Macrófagos/fisiologia , Monócitos/metabolismo , Músculo Esquelético/crescimento & desenvolvimento , Células Satélites de Músculo Esquelético/fisiologia , Células Cultivadas , Fatores Quimiotáticos/genética , Fatores Quimiotáticos/metabolismo , Quimiotaxia , Técnicas de Cocultura , Meios de Cultivo Condicionados , Humanos , Macrófagos/citologia , Dados de Sequência Molecular , Monócitos/citologia , Músculo Esquelético/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Células Satélites de Músculo Esquelético/citologia
8.
Exerc Sport Sci Rev ; 37(1): 18-22, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19098520

RESUMO

Macrophages are necessary for skeletal muscle regeneration after injury. Muscle recruits inflammatory monocytes/macrophages that switch toward an anti-inflammatory profile upon phagocytosis of debris. In vitro, proinflammatory macrophages stimulate myoblast proliferation, whereas anti-inflammatory macrophages stimulate their differentiation. Thus, macrophages are involved in both phases of skeletal muscle regeneration: first, inflammation and cleansing of necrosis, and then myogenic differentiation and tissue repair.


Assuntos
Macrófagos/fisiologia , Músculo Esquelético/fisiologia , Regeneração , Animais , Diferenciação Celular , Humanos , Inflamação/fisiopatologia , Monócitos/fisiologia , Músculo Esquelético/lesões , Mioblastos/citologia , Fenótipo
9.
Methods Mol Biol ; 1891: 19-28, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30414123

RESUMO

The use of an adenoviral vector to transduce cells allows for certain secreted proteins or growth factors to be generated in vivo in eukaryotic cells with accurate posttranslational processing. The use of transduced cells eliminates viral toxicity, allows for targeted expression of the secreted factor at a specific site, and ensures that the therapy will be turned off when the cells are cleared by the organism. Here we describe the delivery system which utilizes cells transduced with a non-replicating adenovirus containing bone morphogenetic protein 2 (BMP-2) in the E1 region of the cassette. With this method of delivery, small amounts of the protein can incite de novo bone formation.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Terapia Baseada em Transplante de Células e Tecidos , Terapia Genética , Adenoviridae/genética , Fosfatase Alcalina , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Genética/métodos , Vetores Genéticos/genética , Humanos , Camundongos , Ratos , Transdução Genética
10.
Mol Biol Cell ; 16(2): 861-70, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15574885

RESUMO

Knowledge on molecular systems involved in myogenic precursor cell (mpc) fusion into myotubes is fragmentary. Previous studies have implicated the a disintegrin and metalloproteinase (ADAM) family in most mammalian cell fusion processes. ADAM12 is likely involved in fusion of murine mpc and human rhabdomyosarcoma cells, but it requires yet unknown molecular partners to launch myogenic cell fusion. ADAM12 was shown able to mediate cell-to-cell attachment through binding alpha9beta1 integrin. We report that normal human mpc express both ADAM12 and alpha9beta1 integrin during their differentiation. Expression of alpha9 parallels that of ADAM12 and culminates at time of fusion. alpha9 and ADAM12 coimmunoprecipitate and participate to mpc adhesion. Inhibition of ADAM12/alpha9beta1 integrin interplay, by either ADAM12 antisense oligonucleotides or blocking antibody to alpha9beta1, inhibited overall mpc fusion by 47-48%, with combination of both strategies increasing inhibition up to 62%. By contrast with blockade of vascular cell adhesion molecule-1/alpha4beta1, which also reduced fusion, exposure to ADAM12 antisense oligonucleotides or anti-alpha9beta1 antibody did not induce detachment of mpc from extracellular matrix, suggesting specific involvement of ADAM12-alpha9beta1 interaction in the fusion process. Evaluation of the fusion rate with regard to the size of myotubes showed that both ADAM12 antisense oligonucleotides and alpha9beta1 blockade inhibited more importantly formation of large (> or =5 nuclei) myotubes than that of small (2-4 nuclei) myotubes. We conclude that both ADAM12 and alpha9beta1 integrin are expressed during postnatal human myogenic differentiation and that their interaction is mainly operative in nascent myotube growth.


Assuntos
Diferenciação Celular , Integrinas/metabolismo , Proteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Desenvolvimento Muscular , Músculo Esquelético/embriologia , Proteínas ADAM , Proteína ADAM12 , Anticorpos Bloqueadores/farmacologia , Adesão Celular , Proliferação de Células , Células Cultivadas , Interações Medicamentosas , Eletroforese em Gel de Poliacrilamida , Fluoresceína-5-Isotiocianato , Imunofluorescência , Corantes Fluorescentes , Humanos , Immunoblotting , Indóis , Integrinas/antagonistas & inibidores , Integrinas/efeitos dos fármacos , Integrinas/genética , Cinética , Fusão de Membrana/efeitos dos fármacos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/efeitos dos fármacos , Metaloendopeptidases/genética , Microscopia Confocal , Fibras Musculares Esqueléticas/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Testes de Precipitina , Propídio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rodaminas
11.
Stem Cells Transl Med ; 6(4): 1109-1119, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28198109

RESUMO

Studies presented here, using a murine model of bone morphogenetic protein type 2 (BMP2)-induced heterotopic ossification (HO) show that the protein initiates HO by signaling through progenitors in the endoneurium of peripheral nerves. In the mouse, these cells were identified in the endoneurium one day after BMP2 induction using antibody against phosphoSMAD (PS) 1, 5, and 8. Studies conducted in a tracking mouse that contains a tamoxifen-regulated Wnt1-Cre recombinase crossed with a td Tomato red (TR) reporter (Wnt1CreErt :Ai9Tm) confirmed their neural origin. In this model both BMP2 induction and tamoxifen are absolutely required to induce TR. SP7+ (osterix+ )TR+ cells were found in the endoneurium on day 1 and associated with bone on day 7. Quantification of TR+ and TR- cells isolated by fluorescence-activated cell sorting showed that all SP7+ cells were found in the TR+ population, whereas only about 80% of the TR+ cells expressed SP7. Pre-chondrocytes (Sox 9+ ) and transient brown fat (tBAT, UCP1+ ) also coexpressed TR, suggesting that the progenitor in nerves is multi-potential. The endoneurium of human nerves near the site of HO contained many PS+ cells, and SP7+ cells were found in nerves and on bone in tissue from patients with HO. Control tissues and nerves did not contain these PS+ and SP7+ cells. Some osteoblasts on bone from patients with HO were positive for PS, suggesting the continued presence of BMP during bone formation. The data suggests that the progenitors for HO are derived from the endoneurium in both the mouse model of HO and in humans with HO. Stem Cells Translational Medicine 2017;6:1109-1119.


Assuntos
Ossificação Heterotópica/metabolismo , Nervos Periféricos/citologia , Nervos Periféricos/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Proteína Morfogenética Óssea 2/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Ferimentos e Lesões/metabolismo
12.
Tissue Eng Part A ; 23(5-6): 177-184, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27967655

RESUMO

Gene therapy approaches have been difficult to implement due to pre-existing immunity against the virus used for delivery. To circumvent this problem, a cell-based approach was developed that avoided the use of free virus within the animal. However, even cells transduced in vitro with E1- to E3-deleted adenovirus encoding bone morphogenetic protein 2 (AdBMP2) resulted in the production of virus-neutralizing antibodies in mice. Furthermore, when mice received an intramuscular injection of nonencoding adenovirus (AdEmpty)-transduced cells, AdBMP2-transduced cells were unable to launch bone formation when an intramuscular injection of these BMP2-producing cells was delivered 1 week later. This phenomenon was not observed in NOD/SCID mice, and could be overcome in C57BL/6 mice by encapsulating the adenovirus-transduced cells in a nondegradable hydrogel poly(ethylene glycol) diacrylate (PEGDA). Data collectively suggest that PEGDA hydrogel encapsulation of AdBMP2-transduced cells prevents pre-existing immunity from suppressing BMP2-induced bone formation.


Assuntos
Adenoviridae , Proteína Morfogenética Óssea 2/imunologia , Células Imobilizadas , Fibroblastos , Hidrogéis/química , Polietilenoglicóis/química , Transdução Genética , Animais , Proteína Morfogenética Óssea 2/genética , Células Imobilizadas/imunologia , Células Imobilizadas/transplante , Fibroblastos/imunologia , Fibroblastos/transplante , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
13.
J Orthop Res ; 34(11): 1894-1904, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26919547

RESUMO

Extremity amputation or traumatic injury can often lead to the formation of heterotopic ossification (HO). Studies to induce HO in rat muscle using cell-based gene therapy show that this process appears to be location dependent. In the present study, HO was induced in mice and rats through injection of immunologically matched cells transduced with either a replication-defective adenovirus possessing bone morphogenetic protein 2 (BMP2) or an empty adenovirus vector (control). Injection in rat near the skeletal bone resulted in HO, whereas cells injected into the same muscle group but distal from the bone did not result in bone formation. When cells were injected in the same limb at both locations at the same time, HO was formed at both sites. Characterization of the bone formation in rats versus mice demonstrated that different sources of osteogenic progenitors were involved, which may account for the location dependent bone formation observed in the rat. Further experimentation has shown that a potential reason for this difference may be the inability of rat to activate matrix metalloproteinase 9 (MMP9), an essential protease in mice necessary for recruitment of progenitors. Inhibition of active MMP9 in mice led to a significant decrease in HO. The studies reported here provide insight into the mechanisms and pathways leading to bone formation in different animals and species. It appears that not all animal models are appropriate for testing HO therapies, and our studies also challenge the conventional wisdom that larger animal models are better for testing treatments affecting bone. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1894-1904, 2016.


Assuntos
Metaloproteinase 9 da Matriz/fisiologia , Ossificação Heterotópica , Adenoviridae , Animais , Células Cultivadas , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Humanos , Transplante de Células-Tronco Mesenquimais , Camundongos Endogâmicos C57BL , Ratos Nus , Ratos Wistar
14.
Cardiovasc Res ; 58(2): 444-50, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12757878

RESUMO

OBJECTIVE: Transplantation of skeletal myogenic precursor cells (mpc) into the myocardium using a non-surgical procedure. METHODS: Closed-chest mpc transplantation was assessed in pigs using the NOGA-Biosense device allowing both electromechanical mapping of the left ventricle (LV), and guided mpc injections through endocardium. RESULTS: We successively established that: (1) adequate preimplantation handling of mpc can be achieved when mpc are kept in 0.1% serum albumin-containing medium until implantation; (2) mpc are neither retained nor destroyed in the catheter or the needle and their passage does not affect their survival, growth and differentiation; (3) large numbers of autologous mpc can be actually transplanted in the LV myocardium by transendocardial route, as assessed by post-mortem examination of pigs injected with iron-loaded mpc; (4) cell injection into the myocardium does not induce conspicuous cell mortality since more than 80% of mpc recovered from LV tissue are alive 15 min after injection; (5) mpc injections can be guided into circumscribed LV targets such as infarcted areas, as assessed by comparison of map injection sites with location of iron-loaded mpc at post-mortem examination of LV myocardium. CONCLUSION: This new approach may pave the way for a large spectrum of cell therapies targeting myocardial diseases.


Assuntos
Insuficiência Cardíaca/cirurgia , Mioblastos Esqueléticos/transplante , Infarto do Miocárdio/cirurgia , Animais , Técnicas de Cultura de Células/métodos , Sobrevivência Celular , Eletrofisiologia , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração , Injeções , Modelos Animais , Mioblastos Esqueléticos/patologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Suínos , Transplante Autólogo
15.
J Orthop Res ; 31(10): 1597-604, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23832813

RESUMO

Current strategies for bone regeneration after traumatic injury often fail to provide adequate healing and integration. Here, we combined the poly (ethylene glycol) diacrylate (PEGDA) hydrogel with allogeneic "carrier" cells transduced with an adenovirus expressing BMP2. The system is unique in that the biomaterial encapsulates the cells, shielding them and thus suppressing destructive inflammatory processes. Using this system, complete healing of a 5 mm-long femur defect in a rat model occurs in under 3 weeks, through secretion of 100-fold lower levels of protein as compared to doses of recombinant BMP2 protein used in studies which lead to healing in 2-3 months. New bone formation was evaluated radiographically, histologically, and biomechanically at 2, 3, 6, 9, and 12 weeks after surgery. Rapid bone formation bridged the defect area and reliably integrated into the adjacent skeletal bone as early as 2 weeks. At 3 weeks, biomechanical analysis showed the new bone to possess 79% of torsional strength of the intact contralateral femur. Histological evaluation showed normal bone healing, with no infiltration of inflammatory cells with the bone being stable approximately 1 year later. We propose that these osteoinductive microspheres offer a more efficacious and safer clinical option over the use of rhBMP2.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Fraturas do Fêmur/tratamento farmacológico , Consolidação da Fratura/efeitos dos fármacos , Polietilenoglicóis/farmacologia , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Fenômenos Biomecânicos/fisiologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Composição de Medicamentos/métodos , Fraturas do Fêmur/diagnóstico por imagem , Fraturas do Fêmur/fisiopatologia , Fêmur/efeitos dos fármacos , Fêmur/fisiologia , Fibroblastos/citologia , Consolidação da Fratura/fisiologia , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Microesferas , Radiografia , Ratos , Ratos Wistar , Pele/citologia , Células Estromais/citologia
16.
Int J Biomater ; 2012: 861794, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22500171

RESUMO

Autologous bone grafting is the most effective treatment for long-bone nonunions, but it poses considerable risks to donors, necessitating the development of alternative therapeutics. Poly(ethylene glycol) (PEG) microencapsulation and BMP2 transgene delivery are being developed together to induce rapid bone formation. However, methods to make these treatments available for clinical applications are presently lacking. In this study we used mesenchymal stem cells (MSCs) due to their ease of harvest, replication potential, and immunomodulatory capabilities. MSCs were from sheep and pig due to their appeal as large animal models for bone nonunion. We demonstrated that cryopreservation of these microencapsulated MSCs did not affect their cell viability, adenoviral BMP2 production, or ability to initiate bone formation. Additionally, microspheres showed no appreciable damage from cryopreservation when examined with light and electron microscopy. These results validate the use of cryopreservation in preserving the viability and functionality of PEG-encapsulated BMP2-transduced MSCs.

17.
PLoS One ; 6(10): e26410, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22039481

RESUMO

Lifelong, many somatic tissues are replenished by specialized adult stem cells. These stem cells are generally rare, infrequently dividing, occupy a unique niche, and can rapidly respond to injury to maintain a steady tissue size. Despite these commonalities, few shared regulatory mechanisms have been identified. Here, we scrutinized data comparing genes expressed in murine long-term hematopoietic stem cells with their differentiated counterparts and observed that a disproportionate number were members of the developmentally-important, monoallelically expressed imprinted genes. Studying a subset, which are members of a purported imprinted gene network (IGN), we found their expression in HSCs rapidly altered upon hematopoietic perturbations. These imprinted genes were also predominantly expressed in stem/progenitor cells of the adult epidermis and skeletal muscle in mice, relative to their differentiated counterparts. The parallel down-regulation of these genes postnatally in response to proliferation and differentiation suggests that the IGN could play a mechanistic role in both cell growth and tissue homeostasis.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica , Animais , Inativação Gênica , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real
18.
J Neuropathol Exp Neurol ; 69(6): 643-53, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20467328

RESUMO

Muscle inflammation can be a prominent feature in several muscular dystrophies. In dysferlin myopathy, it is mainly composed of macrophages. To understand the origin of inflammation in dysferlin-deficient muscle, we analyzed soluble factors involved in monocyte chemotaxis released by myoblasts and myotubes from control and dysferlinopathy patients using a transwell system. Dysferlin-deficient myotubes released more soluble factors involved in monocyte chemotaxis compared with controls (p < 0.001). Messenger RNA microarray analysis showed a 3.2-fold increase of thrombospondin 1 (TSP-1) expression in dysferlin-deficient myotubes. Retrotranscriptasepolymerase chain reaction analysis, ELISA, and immunohistochemistry confirmed these results. Dysferlin mRNA knockdown with short-interfering RNA in normal myogenic cells resulted in TSP-1 mRNA upregulation and increased chemotaxis. Furthermore, monocyte chemotaxis was decreased when TSP-1 was blocked by specific antibodies. In muscle biopsies from dysferlinopathy patients, TSP-1 expression was increased in muscle fibers but not in biopsies of patientswith other myopathies with inflammation; TSP-1 was seen in some macrophages in all samples analyzed. Taken together, the data demonstrate that dysferlin-deficient muscle upregulates TSP-1 in vivoand in vitro and indicate that endogenous chemotactic factors arecrucial to the sustained inflammatory process observed in dysferlinopathies.


Assuntos
Inflamação/genética , Inflamação/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Trombospondina 1/metabolismo , Adulto , Western Blotting , Movimento Celular/fisiologia , Células Cultivadas , Disferlina , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Inflamação/patologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Macrófagos/patologia , Masculino , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/patologia , Distrofias Musculares/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trombospondina 1/genética
19.
J Cell Sci ; 119(Pt 12): 2497-507, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16720640

RESUMO

The mechanisms underlying stromal cell supportive functions are incompletely understood but probably implicate a mixture of cytokines, matrix components and cell adhesion molecules. Skeletal muscle uses recruited macrophages to support post-injury regeneration. We and others have previously shown that macrophages secrete mitogenic factors for myogenic cells. Here, we focused on macrophage-elicited survival signals. We demonstrated that: (1) macrophage influx is temporally correlated with the disappearance of TUNEL-positive apoptotic myogenic cells during post-injury muscle regeneration in mice; (2) direct cell-cell contacts between human macrophages and myogenic cells rescue myogenic cells from apoptosis, as assessed by decreased annexin V labelling and caspase-3 activity, and by increased DIOC-6 staining, Bcl-2 expression and phosphorylation of Akt and ERK1/2 survival pathways; (3) four pro-survival cell-cell adhesion molecular systems detected by DNA macroarray are expressed by macrophages and myogenic cells in vitro and in vivo - VCAM-1-VLA-4, ICAM-1-LFA-1, PECAM-1-PECAM-1 and CX3CL1-CX3CR1; (4) macrophages deliver anti-apoptotic signals through all four adhesion systems, as assessed by functional analyses with blocking antibodies; and (5) macrophages more strongly rescue differentiated myotubes, which must achieve adhesion-induced stabilisation of their structure to survive. Macrophages could secure these cells until they establish final association with the matrix.


Assuntos
Apoptose/fisiologia , Moléculas de Adesão Celular/fisiologia , Macrófagos/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Mioblastos/fisiologia , Animais , Adesão Celular/fisiologia , Membrana Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Venenos Elapídicos/administração & dosagem , Citometria de Fluxo , Humanos , Técnicas In Vitro , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Células Musculares/citologia , Células Musculares/fisiologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA