Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Vet Surg ; 50(5): 1107-1116, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33709467

RESUMO

OBJECTIVE: To determine the feasibility of umbilical cord-derived mesenchymal stem cell (UC-MSC) transplantation into the cervical spinal cord of horses by using fluoroscopy with or without endoscopic guidance and to evaluate the neurological signs and tissue reaction after injection. STUDY DESIGN: Experimental study. ANIMALS: Eight healthy adult horses with no clinical signs of neurological disease. METHODS: After cervical ventral interbody fusion (CVIF), ten million fluorescently labeled allogeneic UC-MSC were injected into the spinal cord under endoscopic and fluoroscopic guidance (n = 5) or fluoroscopic guidance only (n = 3). Postoperative neurological examinations were performed, and horses were humanely killed 48 hours (n = 4) or 14 days (n = 4) postoperatively. Spinal tissues were examined after gross dissection and with bright field and fluorescent microscopy. RESULTS: Needle endoscopy of the cervical canal by ventral approach was associated with intraoperative spinal cord puncture (2/5) and postoperative ataxia (3/5). No intraoperative complications occurred, and one (1/3) horse developed ataxia with cell transplantation under fluoroscopy alone. Umbilical cord-derived MSC were associated with small vessels and detected up to 14 days in the spinal cord. Demyelination was observed in six of eight cases. CONCLUSION: Fluoroscopically guided intramedullary UC-MSC transplantation during CVIF avoids spinal cord trauma and decreases risk of ataxia from endoscopy. Umbilical cord-derived MSC persist in the spinal cord for up to 14 days. Cell injection promotes angiogenesis and induces demyelination of the spinal tissue. CLINICAL SIGNIFICANCE: Umbilical cord-derived MSC transplantation into the spinal cord during CVIF without endoscopy is recommended for future evaluation of cell therapy in horses affected by cervical vertebral compressive myelopathy.


Assuntos
Vértebras Cervicais/cirurgia , Doenças dos Cavalos/cirurgia , Transplante de Células-Tronco Mesenquimais/veterinária , Compressão da Medula Espinal/veterinária , Fusão Vertebral/veterinária , Animais , Ataxia/prevenção & controle , Ataxia/veterinária , Endoscopia/efeitos adversos , Endoscopia/veterinária , Estudos de Viabilidade , Feminino , Fluoroscopia , Cavalos , Masculino , Complicações Pós-Operatórias/veterinária , Compressão da Medula Espinal/cirurgia , Fusão Vertebral/métodos
2.
Biopreserv Biobank ; 18(2): 73-81, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31904273

RESUMO

Background: The growing interest in mesenchymal stromal cells (MSCs) in equine medicine, together with the development of MSC biobanking for allogeneic use, raises concerns about biosafety of such products. MSCs derived from umbilical cord (UC) carry an inherent risk of contamination by environmental conditions and vertical transmission of pathogens from broodmares. There is yet no report in the scientific literature about horses being contaminated by infected MSC products, and no consensus about systematic infectious screening of umbilical cord-derived mesenchymal stromal cells (UC-MSCs) to ensure microbiological safety of therapeutic products. Objectives: To develop a standard protocol to ensure UC-MSC microbiological safety and to assess the risk of vertical transmission of common intracellular pathogens from broodmares to paired UC-MSCs. Study Design and Methods: Eighty-four UC and paired peripheral maternal blood (PMB) samples were collected between 2014 and 2016. Sterility was monitored by microbiological control tests. Maternal contamination was tested by systematical PMB PCR screening for 14 pathogens and a Coggins test. In case of a PCR-positive result regarding one or several pathogen(s) in PMB, a PCR analysis for the detected pathogen(s) was then conducted on the associated UC-MSCs. Results: Ten out of 84 UC samples were contaminated upon extraction and 6/84 remained positive in primo culture. The remaining 78/84 paired PMB & UC-MSC samples were evaluated for vertical transmission; 37/78 PMB samples were PCR positive for Equid herpesvirus (EHV)-1, EHV-2, EHV-5, Theileria equi, Babesia caballi, and/or Mycoplasma spp. Hepacivirus was detected in 2/27 cases and Theiler Diseases Associated Virus in 0/27 cases (not performed on all samples due to late addition). All paired UC-MSC samples tested for the specific pathogen(s) detected in PMB were negative (37/37). Main Limitations: More data are needed regarding MSC susceptibility to most pathogens detected in PMB. Conclusions: In-process microbiological controls combined with PMB PCR screening provide a comprehensive assessment of UC-MSC exposure to infectious risk, vertical transmission risk appearing inherently low.


Assuntos
Bactérias/isolamento & purificação , Células-Tronco Mesenquimais/citologia , Piroplasmida/isolamento & purificação , Cordão Umbilical/citologia , Vírus/isolamento & purificação , Animais , Bancos de Espécimes Biológicos , Contenção de Riscos Biológicos , Feminino , Cavalos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/microbiologia , Leucócitos Mononucleares/parasitologia , Células-Tronco Mesenquimais/microbiologia , Células-Tronco Mesenquimais/parasitologia
3.
PLoS One ; 14(8): e0221317, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31465445

RESUMO

The purpose of this prospective study was to evaluate the effects of single and repeated intra-articular administration of allogeneic, umbilical cord-derived, neonatal mesenchymal stem cells (MSC) in horses with lameness due to osteoarthritis (OA) of a metacarpophalangeal joint (MPJ). Twenty-eight horses were included. Horses were divided into two groups. Horses in group MSC1 received an MSC injection at M0 and a placebo injection at M1 (1 month after M0). Horses in group MSC2 received MSC injections at M0 and at M1. Joint injections were performed with a blinded syringe. Clinical assessment was performed by the treating veterinarian at M1, M2 and M6 (2 and 6 months after M0), including lameness evaluation, palpation and flexion of the joint. Radiographic examination of the treated joints was performed at inclusion and repeated at M6. Radiographs were anonymized and assessed by 2 ECVDI LA associate members. Short term safety assessment was performed by owner survey. A 2-month rehabilitation program was recommended to veterinarians. There was a significant improvement of the total clinical score for horses in both groups. There was no significant difference in the total clinical score between groups MSC1 and MSC2 at any time point in the study. There was no significant difference in the total radiographic OA score, osteophyte score, joint space width score and subchondral bone score between inclusion and M6. Owner-detected adverse effects to MSC injection were recorded in 18% of the horses. Lameness caused by OA improved significantly over the 6-month duration of the study after treatment with allogeneic neonatal umbilical cord-derived MSCs combined with 8 weeks rest and rehabilitation. There is no apparent clinical benefit of repeated intra-articular administration of MSCs at a 1-month interval in horses with MPJ OA when compared to the effect of a single injection.


Assuntos
Doenças dos Cavalos/terapia , Cavalos , Transplante de Células-Tronco Mesenquimais , Articulação Metacarpofalângica , Articulação Metatarsofalângica , Osteoartrite/terapia , Aloenxertos , Animais , Feminino , Doenças dos Cavalos/patologia , Doenças dos Cavalos/fisiopatologia , Masculino , Osteoartrite/patologia , Osteoartrite/fisiopatologia
4.
Sci Rep ; 8(1): 13799, 2018 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-30217993

RESUMO

Umbilical cord blood mesenchymal stromal/stem cells (UCB-MSCs) and umbilical cord matrix MSCs (UCM-MSCs) have chondrogenic potential and are alternative sources to standard surgically derived bone marrow or adipose tissue collection for cartilage engineering. However, the majority of comparative studies explore neonatal MSCs potential only on ISCT benchmark assays accounting for some bias in the reproducibility between in vitro and in clinical studies. Therefore, we characterized equine UCB-MSCs and UCM-MSCs and investigated with particular attention their chondrogenesis potential in 3D culture with BMP-2 + TGF-ß1 in normoxia or hypoxia. We carried out an exhaustive characterization of the extracellular matrix generated by both these two types of MSCs after the induction of chondrogenesis through evaluation of hyaline cartilage, hypertrophic and osteogenic markers (mRNA, protein and histology levels). Some differences in hypoxia sensitivity and chondrogenesis were observed. UCB-MSCs differentiated into chondrocytes express an abundant, dense and a hyaline-like cartilage matrix. By contrast, despite their expression of cartilage markers, UCM-MSCs failed to express a relevant cartilage matrix after chondrogenic induction. Both MSCs types also displayed intrinsic differences at their undifferentiated basal status, UCB-MSCs expressing higher levels of chondrogenic markers whereas UCM-MSCs synthesizing higher amounts of osteogenic markers. Our results suggest that UCB-MSCs should be preferred for ex-vivo horse cartilage engineering. How those results should be translated to in vivo direct cartilage regeneration remains to be determined through dedicated study.


Assuntos
Condrogênese/fisiologia , Sangue Fetal/citologia , Cordão Umbilical/citologia , Animais , Proteína Morfogenética Óssea 2/metabolismo , Cartilagem/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/metabolismo , Condrogênese/genética , Colágeno Tipo I/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Sangue Fetal/fisiologia , Cavalos , Cartilagem Hialina/metabolismo , Células-Tronco Mesenquimais/fisiologia , Osteogênese/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Reprodutibilidade dos Testes , Engenharia Tecidual/métodos , Fator de Crescimento Transformador beta1/metabolismo , Cordão Umbilical/fisiologia
5.
Front Vet Sci ; 4: 83, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28642867

RESUMO

OBJECTIVE: Compare the clinical and pressure walkway gait evolution of dogs after a tibial plateau leveling osteotomy (TPLO) for a cranial cruciate ligament rupture (CrCLR) and treatment with either a 1-month course of non-steroidal anti-inflammatory drugs (NSAIDs) or a single postoperative intra-articular (IA) injection of allogeneic neonatal mesenchymal stromal cells (MSCs). STUDY DESIGN: Prospective, double-blinded, randomized, controlled, monocentric clinical study. ANIMALS: Sixteen client-owned dogs. MATERIALS AND METHODS: Dogs with unilateral CrCLR confirmed by arthroscopy were included. Allogeneic neonatal canine MSCs were obtained from fetal adnexa retrieved after C-section performed on healthy pregnant bitches. The dogs were randomly allocated to either the "MSCs group," receiving an IA injection of MSCs after TPLO, followed by placebo for 1 month, or the "NSAIDs group," receiving IA equivalent volume of MSCs vehicle after TPLO, followed by oral NSAID for 1 month. One of the three blinded evaluators assessed the dogs in each group before and after surgery (1, 3, and 6 months). Clinical score and gait and bone healing process were assessed. The data were statistically compared between the two groups for pre- and postoperative evaluations. RESULTS: Fourteen dogs (nine in the MSCs group, five in the NSAIDs group) completed the present study. No significant difference was observed between the groups preoperatively. No local or systemic adverse effect was observed after MSCs injection at any time point considered. At 1 month after surgery, bone healing scores were significantly higher in the MSCs group. At 1, 3, and 6 months after surgery, no significant difference was observed between the two groups for clinical scores and gait evaluation. CONCLUSION: A single IA injection of allogeneic neonatal MSCs could be a safe and valuable postoperative alternative to NSAIDs for dogs requiring TPLO surgery, particularly for dogs intolerant to this class of drugs.

6.
Vet Immunol Immunopathol ; 171: 47-55, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26964717

RESUMO

In veterinary medicine, therapeutic mesenchymal stromal cells (MSC) have been traditionally isolated from adult bone marrow or adipose tissue. Neonatal tissues, normally discarded at birth from all species have become an alternative source of cells for regenerative medicine in the human clinic. These cells have been described as being more primitive, proliferative and immunosuppressive than their adult counterparts. Our objective was to examine if this phenomena holds true in dogs. Little information exists regarding canine neonatal MSC characterisation. In this study, we were able to both isolate, phenotype and assess the differentiation and immunomodulatory properties of MSC from canine foetal adnexa allowing us to compare their characteristics to their more well-known bone marrow (BM) cousins. Neonatal tissues, including amnion (AM), placenta (PL), and umbilical cord matrix (UCM) were collected from 6 canine caesarean sections. Primary cells were expanded in vitro for 5 consecutive passages and their proliferation measured. BM-MSC were isolated from 5 control dogs euthanised from other studies and grown in vitro using an identical protocol. All MSC lines were systematically evaluated for their ability to differentiate into 3 mesodermal lineages (adipocyte, osteocyte and chondrocyte) and phenotyped by cytometry and qPCR. In addition, the enzymatic activity of the key immunomodulatory marker indoleamine 2,3-dioxygenase (IDO) was evaluated for each MSC line. MSC displaying a fibroblastic appearance were successfully grown from all neonatal tissues. PL-MSC exhibited significantly higher proliferation rates than AM- and UCM-MSC (p=0.05). Cytometric analysis showed that all MSC express CD90, CD29, and CD44, while no expression of CD45, CD34 and MHC2 was detected. Molecular profiling showed expression of CD105 and CD73 in all MSC. Low levels of SOX2 mRNA was observed in all MSC, while neither NANOG, nor OCT4 were detected. All MSC differentiate into 3 mesodermal lineages. Following inflammatory stimulation, the activity of the immunomodulatory enzyme IDO was significantly higher in neonatal MSC compared to BM-MSC (p=0.009). Our results show that canine foetal adnexa cells share very similar properties to their adult equivalents but upon stimulation show significantly higher IDO immunomodulatory activity. Further studies will be needed to confirm the potential therapeutic benefits of these cells.


Assuntos
Cães , Células-Tronco Mesenquimais/citologia , Placenta/citologia , Âmnio/citologia , Animais , Antígenos CD/metabolismo , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Separação Celular , Feminino , Imunomodulação , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Células-Tronco Mesenquimais/imunologia , Gravidez , Cordão Umbilical/citologia
7.
Stem Cells Dev ; 18(9): 1369-78, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19327013

RESUMO

Migration of stem/progenitor cells is a crucial event for homing toward tissue where cells need to be renewed. The neurotransmitter gamma-aminobutyric acid (GABA) has been shown to have a crucial role in migration of neuronal stem/progenitor cells. Since human umbilical cord blood (HUCB) contains stem/progenitor cells able to generate either neuronal or hematopoietic cells, we evaluated the effect of GABA on this type of cells. While whole fraction of mononuclear cells expressed GABA(A) and GABA(B) receptor subunits (GABA-R), only GABA(B)R subunits were found to be expressed on immature CD133+ cells. Functional experiments revealed that both cell fractions of HUCB were attracted by a gradient of GABA concentration and furthermore were blocked by specific antagonists of GABA(A)R and GABA(B)R bicuculline and saclofen, respectively. Moreover, through GABA(B)R activation the migrating fraction was highly enriched by both hematopoietic progenitors and cells able to generate neuron- like cells in culture. Therefore, GABA is a potent chemoattractant of HUCB stem/progenitor cells specifically through GABA(B)R activation.


Assuntos
Quimiotaxia/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco/citologia , Ácido gama-Aminobutírico/farmacologia , Antígeno AC133 , Antígenos CD/metabolismo , Baclofeno/análogos & derivados , Baclofeno/farmacologia , Bicuculina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Sangue Fetal/citologia , Citometria de Fluxo , Imunofluorescência , Antagonistas GABAérgicos/farmacologia , Antagonistas de Receptores de GABA-A , Antagonistas de Receptores de GABA-B , Expressão Gênica , Glicoproteínas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Neurônios/citologia , Neurônios/metabolismo , Peptídeos/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo
8.
Stem Cells Dev ; 17(5): 1005-16, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18811243

RESUMO

Previous studies described that neurons could be generated in vitro from human umbilical cord blood cells. However, there are few data concerning their origin. Notably, cells generating neurons are not well characterized. The present study deals with the origin of cord blood cells generating neurons and mechanisms allowing the neuronal differentiation. We studied neuronal markers of both total fractions of cord blood and stem/progenitor cord blood cells before and after selections and cultures. We also compared neuronal commitment of cord blood cells to that observed for the neuronal cell line SK-N-BE(2). Before cultures, neuronal markers are found within the total fraction of cord blood cells. In CD133+ stem/progenitor cell fraction only immature neuronal markers are detected. However, CD133+ cells are unable to give rise to neurons in cultures, whereas this is achieved when total fraction of cord blood cells is used. In fact, mature functional neurons can be generated from CD133+ cells only in cell-to-cell close contact with either CD133- fraction or a neurogenic epithelium. Furthermore, since CD133+ fraction is heterogenous, we used several selections to precisely identify the phenotype of cord blood-derived neuronal stem/progenitor cells. Results reveal that only CD34- cells from CD133+ fraction possess neuronal potential. These data show the phenotype of cord blood neuronal stem/progenitor cells and the crucial role of direct cell-to-cell contact to achieve their commitment. Identifying the neuron supporting factors may be beneficial to the use of cord blood neuronal stem/progenitor cells for regenerative medicine.


Assuntos
Antígenos CD34/metabolismo , Antígenos CD/metabolismo , Comunicação Celular , Sangue Fetal/citologia , Glicoproteínas/metabolismo , Neurônios/citologia , Peptídeos/metabolismo , Células-Tronco/citologia , Antígeno AC133 , Diferenciação Celular , Linhagem Celular , Humanos , Frações Subcelulares/metabolismo
9.
FASEB J ; 16(12): 1685-7, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12206990

RESUMO

Induction of apoptosis by DNA-damaging agents such as 1-beta-D-arabinofuranosylcytosine (Ara-C) includes the activation of Lyn protein tyrosine kinase. We have previously established that Ara-C-induced activation of Lyn results in its binding to a neutral sphingomyelinase (SMase) and is requisite for its stimulation and the induction of apoptosis in U937 cells. However, the spacio-temporal organization of these events is unclear. This study demonstrates that part of the total cellular SMase activity is sequestered in sphingomyelin-enriched plasma membrane microdomains (rafts). Under Ara-C and daunorubicin (DNR) treatment, Lyn is rapidly activated and translocated into rafts. The compartmentalization of Lyn (as well as neutral SMase activation and apoptosis) induced by these drugs was blocked by the tyrosine kinase inhibitor herbimycin A and raft disruption. In conclusion, this study establishes that DNA-damaging agents such as Ara-C and DNR rapidly induce Lyn activation and its translocation into membrane rafts. This, in turn leads to neutral SMase activation and raft-associated sphingomyelin hydrolysis with the concomitant generation of the proapoptotic lipid second messenger, ceramide. The apparent topological partitioning between DNA damage and apoptosis signaling (integrated into specialized plasma membrane domains) is discussed.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Membrana Celular/efeitos dos fármacos , Citarabina/farmacologia , Daunorrubicina/farmacologia , Esfingomielina Fosfodiesterase/metabolismo , Quinases da Família src/metabolismo , Benzoquinonas , Transporte Biológico/efeitos dos fármacos , Membrana Celular/enzimologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Lactamas Macrocíclicas , Ligação Proteica/efeitos dos fármacos , Quinonas/farmacologia , Rifabutina/análogos & derivados , Células U937 , Quinases da Família src/antagonistas & inibidores
10.
Blood ; 100(4): 1294-301, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12149210

RESUMO

Previous studies demonstrated that Kit activation confers radioprotection. However, the mechanism by which Kit signaling interferes with cellular response to ionizing radiation (IR) has not been firmly established. Based on the role of the sphingomyelin (SM) cycle apoptotic pathway in IR-induced apoptosis, we hypothesized that one of the Kit signaling components might inhibit IR-induced ceramide production or ceramide-induced apoptosis. Results show that, in both Ba/F3 and 32D murine cell lines transfected with wild-type c-kit, stem cell factor (SCF) stimulation resulted in a significant reduction of IR-induced apoptosis and cytotoxicity, whereas DNA repair remained unaffected. Moreover, SCF stimulation inhibited IR-induced neutral sphingomyelinase (N-SMase) stimulation and ceramide production. The SCF inhibitory effect on SM cycle was not influenced by wortmannin, a phosphoinositide-3 kinase (PI3K) inhibitor. The SCF protective effect was maintained in 32D-KitYF719 cells in which the PI3K/Akt signaling pathway is abolished due to mutation in Kit docking site for PI3K. In contrast, phospholipase C gamma (PLC gamma) inhibition by U73122 totally restored IR-induced N-SMase stimulation, ceramide production, and apoptosis in Kit-activated cells. Moreover, SCF did not protect 32D-KitYF728 cells (lacking a functional docking site for PLC gamma 1), from IR-induced SM cycle. Finally, SCF-induced radioprotection of human CD34(+) bone marrow cells was also inhibited by U73122. Altogether, these results suggest that SCF radioprotection is due to PLC gamma 1-dependent negative regulation of IR-induced N-SMase stimulation. Beyond the scope of Kit-expressing cells, it suggests that PLC gamma 1 status could greatly influence the post-DNA damage cellular response to IR, and perhaps, to other genotoxic agents.


Assuntos
Ceramidas/metabolismo , Isoenzimas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Protetores contra Radiação , Transdução de Sinais , Esfingomielinas/metabolismo , Fator de Células-Tronco/fisiologia , Fosfolipases Tipo C/metabolismo , Animais , Apoptose/efeitos da radiação , Linfócitos B/metabolismo , Morte Celular/efeitos da radiação , Linhagem Celular , Radioisótopos de Cobalto , Reparo do DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Expressão Gênica , Humanos , Indóis , Isoenzimas/antagonistas & inibidores , Cinética , Camundongos , Microscopia de Fluorescência , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama , Proteínas Proto-Oncogênicas c-kit/genética , Pirrolidinonas/farmacologia , Esfingomielina Fosfodiesterase/metabolismo , Fator de Células-Tronco/farmacologia , Transfecção , Fosfolipases Tipo C/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA