Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Acta Biomater ; 102: 458-467, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31783141

RESUMO

Engineering of materials consisting of hypertrophic cartilage, as physiological template for de novo bone formation through endochondral ossification (ECO), holds promise as a new class of biological bone substitutes. Here, we assessed the efficiency and reproducibility of bone formation induced by the combination of ceramic granules with fractionated human adipose tissue ("nanofat"), followed by in vitro priming to hypertrophic cartilage. Human nanofat was mixed with different volumetric ratios of ceramic granules (0.2-1 mm) and cultured to sequentially induce proliferation (3 weeks), chondrogenesis (4 weeks), and hypertrophy (2 weeks). The resulting engineered constructs were implanted ectopically in nude mouse. The presence of ceramic granules regulated tissue formation, both in vitro and in vivo. In particular, their dispersion in nanofat at a ratio of 1:16 led to significantly increased cell number and glycosaminoglycan accumulation in vitro, as well as amount and inter-donor reproducibility of bone formation in vivo. Our findings outline a strategy for efficient utilization of nanofat for bone regeneration in an autologous setting, which should now be tested at an orthotopic site. STATEMENT OF SIGNIFICANCE: In this study, we assessed the efficiency and reproducibility of bone formation by a combination of ceramic granules and fractionated human adipose tissue, also known as nanofat, in vitro primed into hypertrophic cartilage. The resulting engineered cartilaginous constructs, when implanted ectopically in nude mouse, resulted in bone and bone marrow formation, more reproducibly and strongly that nanofat alone. This project evaluates the impact of ceramic granules on the functionality and chondrogenic differentiation of mesenchymal progenitors inside their native adipose tissue niche and outlines a novel strategy for an efficient application of nanofat for bone regeneration in an autologous setting.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Regeneração Óssea/efeitos dos fármacos , Cartilagem/metabolismo , Cerâmica/uso terapêutico , Hidroxiapatitas/uso terapêutico , Osteogênese/efeitos dos fármacos , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Tecido Adiposo/transplante , Animais , Remodelação Óssea/efeitos dos fármacos , Cartilagem/transplante , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Camundongos Nus , Neovascularização Fisiológica/efeitos dos fármacos , Reprodutibilidade dos Testes , Transplantes
2.
Cell Transplant ; 26(9): 1496-1504, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-29113460

RESUMO

Bone nonunion is a pathological condition in which all bone healing processes have stopped, resulting in abnormal mobility between 2 bone segments. The incidence of bone-related injuries will increase in an aging population, leading to such injuries reaching epidemic proportions. Tissue engineering and cell therapy using mesenchymal stem cells (MSCs) have raised the possibility of implanting living tissue for bone reconstruction. Bone marrow was first proposed as the source of stem cells for bone regeneration. However, as the quantity of MSCs in the bone marrow decreases, the capacity of osteogenic differentiation of bone marrow stem cells is also impaired by the donor's age in terms of reduced MSC replicative capacity; an increased number of apoptotic cells; formation of colonies positive for alkaline phosphatase; and decreases in the availability, growth potential, and temporal mobilization of MSCs for bone formation in case of fracture. Adipose-derived stem cells (ASCs) demonstrate several advantages over those from bone marrow, including a less invasive harvesting procedure, a higher number of stem cell progenitors from an equivalent amount of tissue harvested, increased proliferation and differentiation capacities, and better angiogenic and osteogenic properties in vivo. Subcutaneous native adipose tissue was not affected by the donor's age in terms of cellular senescence and yield of ASC isolation. In addition, a constant mRNA level of osteocalcin and alkaline phosphatase with a similar level of matrix mineralization of ASCs remained unaffected by donor age after osteogenic differentiation. The secretome of ASCs was also unaffected by age when aiming to promote angiogenesis by vascular endothelial growth factor (VEGF) release in hypoxic conditions. Therefore, the use of adipose cells for bone tissue engineering is not limited by the donor's age from the isolation of stem cells up to the manufacturing of a complex osteogenic graft.


Assuntos
Tecido Adiposo/citologia , Células-Tronco/citologia , Engenharia Tecidual/métodos , Fatores Etários , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Diabetes Obes Metab ; 19(8): 1061-1070, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28116849

RESUMO

AIMS: To understand better the control of insulin secretion by human ß cells and to identify similarities to and differences from rodent models. METHODS: Dynamic insulin secretion was measured in perifused human islets treated with pharmacological agents of known modes of action. RESULTS: Glucokinase activation (Ro28-1675) lowered the glucose threshold for stimulation of insulin secretion to 1 mmol/L (G1), augmented the response to G3-G5 but not to G8-G15, whereas tolbutamide remained active in G20, which indicates that not all KATP channels were closed by high glucose concentrations. An almost 2-fold greater response to G15 than to supramaximal tolbutamide in G3 or to KCl+diazoxide in G15 vs G3 quantified the contribution of metabolic amplification to insulin secretion. Both disruption (latrunculin-B) and stabilization (jasplakinolide) of microfilaments augmented insulin secretion without affecting metabolic amplification. Tolbutamide-induced insulin secretion was consistently greater in G10 than G3, with a threshold at 1 and maximum at 10 µmol/L tolbutamide in G10, vs 10 and 25 µmol/L in G3. Sulphonylurea effects were thus clearly glucose-dependent. Insulin secretion was also increased by inhibiting K channels other than KATP channels: Kv or BK channels (tetraethylammonium), TASK-1 channels (ML-365) and SK4 channels (TRAM-34). Opening KATP channels with diazoxide inhibited glucose-induced insulin secretion with half maximum inhibitory concentrations of 9.6 and 24 µmol/L at G7 and G15. Blockade of L-type Ca channels (nimodipine) abolished insulin secretion, whereas a blocker of T-type Ca channels (NNC-55-0396) was ineffective at specific concentrations. Blockade of Na channels (tetrodotoxin) did not affect glucose-induced insulin secretion. CONCLUSIONS: In addition to sharing a KATP channel-dependent triggering pathway and a metabolic amplifying pathway, human and rodent ß cells were found to display more similarities than differences in the control of insulin secretion.


Assuntos
Sinalização do Cálcio , Exocitose , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Canais KATP/metabolismo , Modelos Biológicos , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Adulto , Animais , Sinalização do Cálcio/efeitos dos fármacos , Exocitose/efeitos dos fármacos , Feminino , Humanos , Hipoglicemiantes/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Canais KATP/antagonistas & inibidores , Masculino , Moduladores de Transporte de Membrana/farmacologia , Camundongos , Ratos , Especificidade da Espécie , Compostos de Sulfonilureia/farmacologia , Técnicas de Cultura de Tecidos , Doadores de Tecidos
4.
PLoS One ; 11(12): e0168058, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27992567

RESUMO

AIM: Adipose-derived stem cells (ASC) are currently proposed for wound healing in those with type 2 diabetes mellitus (T2DM). Therefore, this study investigated the impact of diabetes on adipose tissue in relation to ASC isolation, proliferation, and growth factor release and the impact of hyperglycemia and low oxygen tension (found in diabetic wounds) on dermal fibroblasts, keratinocytes, and ASC in vitro. METHODS: Different sequences of hypoxia and hyperglycemia were applied in vitro to ASC from nondiabetic (n = 8) or T2DM patients (n = 4) to study cell survival, proliferation, and growth factor release. Comparisons of dermal fibroblasts (n = 8) and keratinocytes (primary lineage) were made. RESULTS: No significant difference of isolation and proliferation capacities was found in ASC from nondiabetic and diabetic humans. Hypoxia and hyperglycemia did not impact cell viability and proliferation. Keratinocyte Growth Factor release was significantly lower in diabetic ASC than in nondiabetic ASC group in each condition, while Vascular Endothelial Growth Factor release was not affected by the diabetic origin. Nondiabetic ASC exposition to hypoxia (0.1% oxygen) combined with hyperglycemia (25mM glucose), resulted in a significant increase in VEGF secretion (+64%, p<0.05) with no deleterious impact on KGF release in comparison to physiological conditions (5% oxygen and 5 mM glucose). Stromal cell-Derived Factor-1α (-93%, p<0.001) and KGF (-20%, p<0.05) secretion by DF decreased in these conditions. CONCLUSIONS: A better profile of growth factor secretion (regarding wound healing) was found in vitro for ASC in hyperglycemia coupled with hypoxia in comparison to dermal fibroblasts and keratinocytes. Interestingly, ASC from T2DM donors demonstrated cellular growth rates and survival (in hypoxia and hyperglycemic conditions) similar to those of healthy ASC (from normoglycemic donors); however, KGF secretion was significantly depleted in ASC obtained from T2DM patients. This study demonstrated the impact of diabetes on ASC for regenerative medicine and wound healing.


Assuntos
Tecido Adiposo/citologia , Diabetes Mellitus Tipo 2/patologia , Fibroblastos/citologia , Glucose/farmacologia , Queratinócitos/citologia , Células-Tronco/citologia , Tecido Adiposo/metabolismo , Adulto , Idoso , Hipóxia Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Fibroblastos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Queratinócitos/metabolismo , Masculino , Pessoa de Meia-Idade , Células-Tronco/metabolismo , Cicatrização , Adulto Jovem
5.
Stem Cells Transl Med ; 5(11): 1575-1579, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27400794

RESUMO

: It is important to standardize methods to quantify the purity of adipose tissue-derived cells for regenerative medicine. We developed a simple and robust tool to discriminate fibroblasts and adipose stem cells (ASCs) by testing release of specific growth factors. ASCs and dermal fibroblasts (DFs) were isolated from human donors (n = 8). At passage 4, cultures were prepared with progressive ASC/DF ratios of 100%/0%, 75%/25%, 50%/50%, 25%/75%, and 0%/100% for each donor and incubated in hypoxic chambers at 0.1% and 5% O2 and hyperglycemia at 1.0 and 4.5 g/l. After incubation for 24 hours, cell survival, proliferation, and growth factor release (vascular endothelial growth factor [VEGF], hepatocyte growth factor [HGF], insulin-like growth factor 1 [IGF-1], stromal cell-derived factor 1α [SDF-1α], and basic fibroblast growth factor [bFGF]) were assessed for each condition. The proliferation and viability of ASCs and DFs were not impacted by the oxygen tension conditions. No significant difference in HGF, IGF-1, bFGF, and keratinocyte growth factor secretome was found across the various ASC/DF ratios. Interestingly, a negative relation for VEGF secretion was found when ASCs were contaminated by fibroblasts, especially when cells were exposed to 4.5 g/l glucose and 0.1% O2 (R = -0.521; p < .001). In contrast, secretion of SDF-1α was positively correlated with the fibroblast ratio, more prominently in low glucose and low oxygen tension (r = .657; p < .001). Above and beyond these previously unreported metabolic features, these results (a) allow us to discriminate fibroblasts and ASCs specifically and (b) allow new tools be developed for the rapid testing (a response within 24 hours) for the release of ASC-based therapies. SIGNIFICANCE: In order to provide direction to academia, industry, and regulatory authorities regarding purity assessment for adipose tissue-derived cells, this report describes a simple tool to facilitate development of international standards based on reproducible parameters and endpoints that may systematize cellular products across boundaries and accelerate the delivery of safe and effective adipose stem cell (ASC)-based tools to the medical community and the patients it serves. This tool (a) can discriminate specifically fibroblasts and ASCs and (b) can be rapidly implemented and performed before the release of the ASC-based therapy (a response within 24 hours).

6.
Medicine (Baltimore) ; 94(50): e2220, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26683933

RESUMO

Long bone nonunion in the context of congenital pseudarthrosis or carcinologic resection (with intercalary bone allograft implantation) is one of the most challenging pathologies in pediatric orthopedics. Autologous cancellous bone remains the gold standard in this context of long bone nonunion reconstruction, but with several clinical limitations. We then assessed the feasibility and safety of human autologous scaffold-free osteogenic 3-dimensional (3D) graft (derived from autologous adipose-derived stem cells [ASCs]) to cure a bone nonunion in extreme clinical and pathophysiological conditions. Human ASCs (obtained from subcutaneous adipose tissue of 6 patients and expanded up to passage 4) were incubated in osteogenic media and supplemented with demineralized bone matrix to obtain the scaffold-free 3D osteogenic structure as confirmed in vitro by histomorphometry for osteogenesis and mineralization. The 3D "bone-like" structure was finally transplanted for 3 patients with bone tumor and 3 patients with bone pseudarthrosis (2 congenital, 1 acquired) to assess the clinical feasibility, safety, and efficacy. Although minor clones with structural aberrations (aneuploidies, such as tri or tetraploidies or clonal trisomy 7 in 6%-20% of cells) were detected in the undifferentiated ASCs at passage 4, the osteogenic differentiation significantly reduced these clonal anomalies. The final osteogenic product was stable, did not rupture with forceps manipulation, did not induce donor site morbidity, and was easily implanted directly into the bone defect. No acute (<3 mo) side effects, such as impaired wound healing, pain, inflammatory reaction, and infection, or long-term side effects, such as tumor development, were associated with the graft up to 4 years after transplantation. We report for the first time that autologous ASC can be fully differentiated into a 3D osteogenic-like implant without any scaffold. We demonstrated that this engineered tissue can safely promote osteogenesis in extreme conditions of bone nonunions with minor donor site morbidity and no oncological side effects.


Assuntos
Adipócitos/citologia , Pseudoartrose/terapia , Engenharia Tecidual/métodos , Adolescente , Neoplasias Ósseas/patologia , Neoplasias Ósseas/cirurgia , Diferenciação Celular , Criança , Estudos de Viabilidade , Feminino , Humanos , Masculino , Osteossarcoma/patologia , Osteossarcoma/cirurgia , Pseudoartrose/etiologia , Pseudoartrose/patologia , Células-Tronco , Alicerces Teciduais
7.
PLoS One ; 10(12): e0144914, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26659378

RESUMO

A lack of oxygen is classically described as a major cause of impaired wound healing in diabetic patients. Even if the role of oxygen in the wound healing process is well recognized, measurement of oxygen levels in a wound remains challenging. The purpose of the present study was to assess the value of electron paramagnetic resonance (EPR) oximetry to monitor pO2 in wounds during the healing process in diabetic mouse models. Kinetics of wound closure were carried out in streptozotocin (STZ)-treated and db/db mice. The pO2 was followed repeatedly during the healing process by 1 GHz EPR spectroscopy with lithium phthalocyanine (LiPc) crystals used as oxygen sensor in two different wound models: a full-thickness excisional skin wound and a pedicled skin flap. Wound closure kinetics were dramatically slower in 12-week-old db/db compared to control (db/+) mice, whereas kinetics were not statistically different in STZ-treated compared to control mice. At the center of excisional wounds, measurements were highly influenced by atmospheric oxygen early in the healing process. In pedicled flaps, hypoxia was observed early after wounding. While reoxygenation occurred over time in db/+ mice, hypoxia was prolonged in the diabetic db/db model. This observation was consistent with impaired healing and microangiopathies observed using intravital microscopy. In conclusion, EPR oximetry using LiPc crystals as the oxygen sensor is an appropriate technique to follow wound oxygenation in acute and chronic wounds, in normal and diabetic animals. Nevertheless, the technique is limited for measurements in pedicled skin flaps and cannot be applied to excisional wounds in which diffusion of atmospheric oxygen significantly affects the measurements.


Assuntos
Diabetes Mellitus Experimental/complicações , Oxigênio/metabolismo , Cicatrização , Animais , Glicemia/análise , Estudos de Casos e Controles , Diabetes Mellitus Experimental/induzido quimicamente , Modelos Animais de Doenças , Espectroscopia de Ressonância de Spin Eletrônica , Microscopia Intravital , Masculino , Camundongos , Camundongos Obesos , Oximetria , Dermatopatias/complicações , Dermatopatias/patologia
8.
PLoS One ; 10(10): e0139566, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26485394

RESUMO

Based on immunomodulatory, osteogenic, and pro-angiogenic properties of adipose-derived stem cells (ASCs), this study aims to assess the safety and efficacy of ASC-derived cell therapies for clinical indications. Two autologous ASC-derived products were proposed to 17 patients who had not experienced any success with conventional therapies: (1) a scaffold-free osteogenic three-dimensional graft for the treatment of bone non-union and (2) a biological dressing for dermal reconstruction of non-healing chronic wounds. Safety was studied using the quality control of the final product (genetic stability, microbiological/mycoplasma/endotoxin contamination) and the in vivo evaluation of adverse events after transplantation. Feasibility was assessed by the ability to reproducibly obtain the final ASC-based product with specific characteristics, the time necessary for graft manufacturing, the capacity to produce enough material to treat the lesion, the surgical handling of the graft, and the ability to manufacture the graft in line with hospital exemption regulations. For 16 patients (one patient did not undergo grafting because of spontaneous bone healing), in-process controls found no microbiological/mycoplasma/endotoxin contamination, no obvious deleterious genomic anomalies, and optimal ASC purity. Each type of graft was reproducibly obtained without significant delay for implantation and surgical handling was always according to the surgical procedure and the implantation site. No serious adverse events were noted for up to 54 months. We demonstrated that autologous ASC transplantation can be considered a safe and feasible therapy tool for extreme clinical indications of ASC properties and physiopathology of disease.


Assuntos
Adipócitos/transplante , Regeneração Óssea/fisiologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais , Procedimentos de Cirurgia Plástica/métodos , Adolescente , Adulto , Idoso , Criança , Estudos de Viabilidade , Feminino , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Pessoa de Meia-Idade , Alicerces Teciduais , Transplante Autólogo , Adulto Jovem
9.
Am J Physiol Endocrinol Metab ; 309(7): E640-50, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26264556

RESUMO

The biphasic pattern of glucose-induced insulin secretion is altered in type 2 diabetes. Impairment of the first phase is an early sign of ß-cell dysfunction, but the underlying mechanisms are still unknown. Their identification through in vitro comparisons of islets from diabetic and control subjects requires characterization and quantification of the dynamics of insulin secretion by normal islets. When perifused normal human islets were stimulated with 15 mmol/l glucose (G15), the proinsulin/insulin ratio in secretory products rapidly and reversibly decreased (∼50%) and did not reaugment with time. Switching from prestimulatory G3 to G6-G30 induced biphasic insulin secretion with flat but sustained (2 h) second phases. Stimulation index reached 6.7- and 3.6-fold for the first and second phases induced by G10. Concentration dependency was similar for both phases, with half-maximal and maximal responses at G6.5 and G15, respectively. First-phase response to G15-G30 was diminished by short (30-60 min) prestimulation in G6 (vs. G3) and abolished by prestimulation in G8, whereas the second phase was unaffected. After 1-2 days of culture in G8 (instead of G5), islets were virtually unresponsive to G15. In both settings, a brief return to G3-G5 or transient omission of CaCl2 restored biphasic insulin secretion. Strikingly, tolbutamide and arginine evoked immediate insulin secretion in islets refractory to glucose. In conclusion, we quantitatively characterized the dynamics of glucose-induced insulin secretion in normal human islets and showed that slight elevation of prestimulatory glucose reversibly impairs the first phase, which supports the view that the similar impairment in type 2 diabetic patients might partially be a secondary phenomenon.


Assuntos
Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Adolescente , Adulto , Células Cultivadas , Feminino , Humanos , Secreção de Insulina , Cinética , Masculino , Pessoa de Meia-Idade , Proinsulina/metabolismo , Adulto Jovem
10.
Plast Reconstr Surg ; 136(2): 279-295, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25946602

RESUMO

BACKGROUND: Nonhealing wounds are unable to integrate skin autografts by avascular and fibrotic dermal tissue. Adipose-derived stromal cells can improve the local environment of the wound bed by angiogenesis and immunomodulation. This work aimed to develop a biological dressing made of adipose-derived stromal cells onto a human acellular collagen matrix. METHODS: Adipose-derived stromal cells were isolated from human adipose tissue (n = 8). In vitro, the genetic stability during early and late passages (1, 4, 10, and 16) and vascular endothelial growth factor (VEGF) secretion were assessed. Adipose-derived stromal cell adhesion and spreading on collagen matrix were preliminarily studied. In vivo tumorigenicity, angiogenesis, and tissue oxygenation were assessed after implantation of the construct in nude rats (n = 10). The biological dressing was manufactured and implanted in three patients with chronic wounds. RESULTS: In vitro, aneuploidies, but no clonal transformation, were detected up to late cellular passages. VEGF was secreted more during hypoxia (0.1% oxygen) than during normoxia (21% oxygen). Adipose-derived stromal cells can adhere and spread on the scaffold within 18 to 20 days. No tumor development occurred 3 months after implantation in immunocompromised rats. Vessel counts and tissue oxygenation were higher after adipose-derived stromal cell implantation. In patients, granulation tissue was found (276 percent of vessel density), followed by epithelialization or split-thickness skin engraftment up to 22 months after implantation. CONCLUSIONS: Implantation of adipose-derived stromal cells seeded onto human acellular collagen matrix (biological dressing) represents a promising therapy for nonhealing wounds, offering improvement in dermal angiogenesis and remodeling. This therapy using autologous stromal cells is safe, without significant genetic alterations after in vitro expansion.


Assuntos
Adipócitos/transplante , Curativos Biológicos/estatística & dados numéricos , Colágeno/fisiologia , Neovascularização Fisiológica/fisiologia , Células Estromais/transplante , Ferimentos e Lesões/diagnóstico por imagem , Animais , Doença Crônica , Técnicas de Cocultura/métodos , Humanos , Ratos , Ratos Sprague-Dawley , Regeneração/fisiologia , Sensibilidade e Especificidade , Pele Artificial , Transplante Autólogo , Ultrassonografia , Cicatrização/fisiologia
11.
Acta Orthop Belg ; 80(2): 196-204, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25090792

RESUMO

Bone allografts were used in our department since twenty-five years to reconstruct segmental bone loss and our data were retrospectively reviewed to assess the complications related to the use of a bone allograft. A consecutive series of 128 patients who received a bone allograft was analyzed. The minimal follow-up was 18 months. Fracture, nonunion, infection and explantation were investigated using a multivariate analysis and logistical regression. Kaplan-Meier survival of the allograft was performed, using allograft removal as the end point. Tumour disease was excluded from this study. Patients were followed up for an average 103 months. Bone tumour occurred in 78% of the patients whereas revision arthroplasty was the cause of implantation in 15% of them. Nonunion was the most prevalent complication, occurring in 35% of the grafts. For nonunion occurrence, the type of reconstruction was found to be a significant variable, the intercalary allograft being the most exposed. Primary bone autografting at the anastomotic site was not significant to prevent nonunion. Fracture of the allograft was the second most frequent complication with a prevalence of 16.4%. The length of the allograft and an osteoarticular allograft were two significant variables in that occurrence. Infection of the allograft was present with a rate of 5.4% of patients. Explantation of failed allografts occurred in 30% of them. The duration of the frozen storage of the allograft and the donor age of the allograft were not significant on any local complication occurrence. Bone allografts are a reliable material but a high rate of local complications must be anticipated.


Assuntos
Transplante Ósseo/efeitos adversos , Transplante Ósseo/métodos , Adulto , Aloenxertos , Feminino , Seguimentos , Humanos , Masculino , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/etiologia , Prevalência , Procedimentos de Cirurgia Plástica , Estudos Retrospectivos , Transplante Homólogo
12.
Cell Transplant ; 23(11): 1349-64, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23461890

RESUMO

Insufficient oxygenation can limit the long-term survival of encapsulated islets in subcutaneous tissue. Transplantation of coencapsulated pig islets with adipose or bone marrow mesenchymal stem cells (AMSCs or BM-MSCs, respectively) was investigated with regard to implant vascularization, oxygenation, and diabetes correction in primates. The in vivo impact of MSCs on graft oxygenation and neovascularization was assessed in rats with streptozotocin (STZ)-induced diabetes that were subcutaneously transplanted with islets coencapsulated with AMSCs (n = 8) or BM-MSCs (n = 6). Results were compared to islets encapsulated alone (n = 8). STZ diabetic primates were subcutaneously transplanted with islets coencapsulated with BM-MSCs (n = 4) or AMSCs (n = 6). Recipients were monitored metabolically and immunologically, and neoangiogenesis was assessed on explanted grafts. Results were compared with primates transplanted with islets encapsulated alone (n = 5). The cotransplantation of islets with BM-MSCs or AMSCs in diabetic rats showed significantly higher graft oxygenation than islets alone (3% and 3.6% O2 for islets + BM-MSCs or AMSCs, respectively, vs. 2.2% for islets alone). A significantly better glycated hemoglobin correction (28 weeks posttransplantation) was found for primates transplanted with islets and MSCs (7.4% and 8.1%, respectively) in comparison to islets encapsulated alone (10.9%). Greater neoangiogenesis was found in the periphery of coencapsulated islets and AMSCs in comparison to islets alone (p < 0.01). In conclusion, the coencapsulation of pig islets with MSCs can improve significantly the islets' survival/function in vitro. The coencapsulation of islets with MSCs improves implant oxygenation and neoangiogenesis. However, the cotransplantation of islets with MSCs improves only slightly the long-term function of a subcutaneous bioartificial pancreas in a primate preclinical model.


Assuntos
Órgãos Bioartificiais , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Pâncreas/irrigação sanguínea , Animais , Engenharia Celular/métodos , Feminino , Ilhotas Pancreáticas/metabolismo , Macaca fascicularis , Masculino , Células-Tronco Mesenquimais/metabolismo , Ratos , Ratos Wistar , Suínos , Transplante Heterólogo/métodos
13.
Curr Diab Rep ; 13(5): 745-55, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23959794

RESUMO

Although islet transplantation has demonstrated its potential use in treating type 1 diabetes, this remains limited by the need for daily immunosuppression. Islet encapsulation was then proposed with a view to avoiding any immunosuppressive regimen and related side effects. In order to obtain a standard clinical procedure in terms of safety and reproducibility, two important factors have to be taken into account: the encapsulation design (which determines the graft volume) and the implantation site. Indeed, the implantation site should meet certain requirements: (1) its space must be large enough for the volume of transplanted tissues; (2) there must be proximity to abundant vascularization with a good oxygen supply; (3) there must be real-time access to physiologically representative blood glucose levels; (4) there must be easy access for implantation and the reversibility of the procedure (for safety); and finally, (5) the site should have minimal early inflammatory reaction and promote long-term survival. The aim of this article is to review possible preclinical/clinical implantation sites (in comparison with free islets) for encapsulated islet transplantation as a function of the encapsulation design: macro/microcapsules and conformal coating.


Assuntos
Bioengenharia/métodos , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/citologia , Animais , Células Imobilizadas/citologia , Células Imobilizadas/metabolismo , Ensaios Clínicos como Assunto , Humanos , Implantes Experimentais
14.
Biomaterials ; 34(18): 4428-38, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23507085

RESUMO

For critical size bone defects and bone non-unions, bone tissue engineering using osteoblastic differentiated adipose mesenchymal stem cells (AMSCs) is limited by the need for a biomaterial to support cell transplantation. An osteoblastic three-dimensional autologous graft made of AMSCs (3D AMSC) was developed to solve this issue. This autograft was obtained by supplementing the osteoblastic differentiation medium with demineralized bone matrix. Two surgical models were developed to assess the potential of this 3D osteogenic AMSC autograft. A four-level spinal fusion using polyetheretherketone cages was designed in six pigs to assess the early phase of ossification (8-12 weeks postimplantation). In each pig, four groups were compared: cancellous bone autograft, freeze-dried irradiated cancellous pig bone, 3D AMSC, and an empty cage. A critical size femoral defect (n = 4, bone non-union confirmed 6 months postoperatively) was used to assess the 3D AMSCs' ability to achieve bone fusion. Pigs were followed by CT scan and explanted specimens were analyzed for bone tissue remodeling by micro-CT scan, micro-radiography, and histology/histomorphometry. In the spine fusion model, bone formation with the 3D AMSC was demonstrated by a significant increase in bone content. In the critical-size femoral defect model, the 3D AMSC achieved new bone formation and fusion in a poorly vascularized fibrotic environment. This custom-made 3D osteogenic AMSC autograft is a therapeutic solution for bone non-unions and for critical-size defects.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular , Fêmur/patologia , Células-Tronco Mesenquimais/citologia , Osteogênese , Medicina Regenerativa/métodos , Coluna Vertebral/patologia , Animais , Benzofenonas , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Fraturas não Consolidadas/diagnóstico por imagem , Fraturas não Consolidadas/patologia , Implantes Experimentais , Cetonas/farmacologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Polietilenoglicóis/farmacologia , Polímeros , Implantação de Prótese , Fusão Vertebral , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/efeitos dos fármacos , Sus scrofa , Transplante Autólogo , Microtomografia por Raio-X
15.
Biomaterials ; 34(13): 3279-89, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23375391

RESUMO

Pig adipose mesenchymal stem cells (AMSCs) could be proposed for the improvement of bone substitute. However, these xenogenic cells retain a galactosyl (Gal) epitope that elicits xenorejection. Our work aims to use Gal-Knock-Out (Gal-KO) pig AMSCs to associate cellular immunomodulation, humoral down-elicitation of Gal-KO cells and osteogenic capacity of AMSCs. Human and pig AMSCs were compared for proliferation/differentiation kinetics and bone neoformation in vivo. Humoral reaction against pig Gal+ vs. Gal-KO AMSCs and immunomodulation properties of Gal+ vs. Gal-KO AMSCs were assessed in vitro. Humoral/cellular reactions against Gal+ vs. Gal-KO osteogenic differentiated pig AMSC xenografts were assessed in an immunocompetent rodent model. Expansion/differentiation/bone neoformation was significantly improved with differentiated pig AMSCs compared with human cells. Based on immunohistochemistry and cell-based ELISA, Gal+ AMSCs had higher sensitivity to preformed/induced anti-pig antibodies than Gal-KO AMSCs. In vitro cellular immunomodulation was similar between Gal+ and Gal-KO AMSCs. In vivo, a significant reduction of anti-pig IgG was found at 1 month in rats implanted with Gal-KO AMSCs compared with those implanted with Gal+ AMSCs. Lymphocyte/macrophage infiltration of osteogenic differentiated pig AMSC xenografts was significantly lower at post-operative day (POD) 7 in recipients of Gal-KO vs. Gal+ pig cells. No significant difference was found at POD 28. The combination of the cellular immunomodulation with the Gal-KO phenotype of AMSCs can significantly improve the cellular engraftment of pig osteogenic cells by delaying xenorejection.


Assuntos
Tecido Adiposo/citologia , Regeneração Óssea , Galactose/metabolismo , Técnicas de Inativação de Genes , Células-Tronco Mesenquimais/citologia , Transplante Heterólogo , Animais , Antígenos/imunologia , Diferenciação Celular , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Epitopos/metabolismo , Galactose/imunologia , Humanos , Imunidade Humoral/imunologia , Imunização , Imuno-Histoquímica , Imunomodulação/imunologia , Cinética , Teste de Cultura Mista de Linfócitos , Células-Tronco Mesenquimais/metabolismo , Modelos Animais , Osteogênese , Primatas/imunologia , Ratos , Ratos Wistar , Sus scrofa
16.
Cell Transplant ; 22(11): 2161-73, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23051152

RESUMO

Pig islets demonstrate significantly lower insulin secretion after glucose stimulation than human islets (stimulation index of ∼12 vs. 2 for glucose 1 and 15 mM, respectively) due to a major difference in ß- and α-cell composition in islets (60% and 25% in humans and 90% and 8% in pigs, respectively). This leads to a lower rise in 3',5'-cyclic adenosine monophosphate (cAMP) in pig ß-cells. Since glucagon is the major hormonal effector of cAMP in ß-cells, we modified pig islet structure in vivo to increase the proportion of α-cells per islet and to improve insulin secretion. Selected doses (0, 30, 50, 75, and 100 mg/kg) of streptozotocin (STZ) were intravenously injected in 32 young pigs to assess pancreatic (insulin and glucagon) hormone levels, islet remodeling (histomorphometry for α- and ß-cell proportions), and insulin and glucagon secretion in isolated islets. Endocrine structure and hormonal content of pig islets were compared with those of human islets. The dose of STZ was significantly correlated with reductions in pancreatic insulin content (p< 0.05, r(2) = 0.77) and the proportion of ß-cells (p < 0.05, r(2) = 0.88). A maximum of 50 mg/kg STZ was required for optimal structure remodeling, with an increased proportion of α-cells per islet (26% vs. 48% α-cells per islet for STZ <50 mg/kg vs. >75 mg/kg; p < 0.05) without ß-cell dysfunction. Three months after STZ treatment (30/50 mg/kg STZ), pig islets were isolated and compared with isolated control islets (0 mg/kg STZ). Isolated islets from STZ-treated (30/50 mg/kg) pigs had a higher proportion of α-cells than those from control animals (32.0% vs. 9.6%, respectively, p < 0.05). After in vitro stimulation, isolated islets from STZ-treated pigs demonstrated significantly higher glucagon content (65.4 vs. 21.0 ng/ml, p < 0.05) and insulin release (144 µU/ml) than nontreated islets (59 µU/ml, p < 0.05), respectively. Low-dose STZ (<50 mg/kg) can modify the structure of pig islets in vivo and improve insulin secretion after isolation.


Assuntos
Células Secretoras de Glucagon/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Estreptozocina/toxicidade , Adulto , Animais , Glicemia/análise , Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/anatomia & histologia , Masculino , Pessoa de Meia-Idade , Suínos
17.
Cell Tissue Bank ; 13(3): 487-98, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22718427

RESUMO

With this analysis we would like to raise some issues that emerge as a result of recent evolutions in the burgeoning field of human cells, tissues, and cellular and tissue-based product (HCT/P) transplantation, and this in the light of the current EU regulatory framework. This paper is intended as an open letter addressed to the EU policy makers, who will be charged with the review and revision of the current legislation. We propose some urgent corrections or additions to cope with the rapid advances in biomedical science, an extensive commercialization of HCT/Ps, and the growing expectation of the general public regarding the ethical use of altruistically donated cells and tissues. Without a sound wake-up call, the diverging interests of this newly established 'healthcare' industry and the wellbeing of humanity will likely lead to totally unacceptable situations, like some of which we are reporting here.


Assuntos
Preparações Farmacêuticas/economia , Bancos de Tecidos/economia , Transplante Homólogo/economia , União Europeia , Humanos , Internacionalidade , Transferência de Tecnologia
18.
Transplant Rev (Orlando) ; 26(3): 183-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22000658

RESUMO

Allogeneic islet transplantation has proven difficult because organ shortages are recurrent, several pancreas donors are often needed to treat one diabetic recipient, and the intrahepatic site of islet implantation may not be the most appropriate site. Thus, another source of insulin-producing cells would be beneficial; and pigs represent a possible and viable source for obtaining such cells. Although the use of pig islet grafts appears to be difficult because of the species barrier, recent reports demonstrated that pig islet xenotransplantation can overcome the immunological barrier following strong immunosuppression and function successfully in primates for at least 6 months. Before becoming clinically applicable, however, pig islet xenotransplantation must still overcome the structural and physiological incompatibility between pig donor and human recipient. Researchers agree that it is necessary to produce more preclinical data in the pig-to-primate model before any pig-to-human transplantation of islets can be considered. Therefore, in this review, we provide a summary of the present state of knowledge about pig and human islet compatibility.


Assuntos
Diabetes Mellitus/cirurgia , Transplante das Ilhotas Pancreáticas , Sus scrofa , Transplante Heterólogo , Animais , Glicemia/metabolismo , Diabetes Mellitus/sangue , Humanos , Terapia de Imunossupressão , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/imunologia
19.
World J Gastroenterol ; 18(47): 6885-93, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23322985

RESUMO

Although allogeneic islet transplantation can successfully cure type 1 diabetes, it has limited applicability. For example, organs are in short supply; several human pancreas donors are often needed to treat one diabetic recipient; the intrahepatic site may not be the most appropriate site for islet implantation; and immunosuppressive regimens, which are associated with side effects, are often required to prolong survival of the islet graft. An alternative source of insulin-producing cells would therefore be of major interest. Pigs represent a possible alternative source of beta cells. Grafting of pig islets may appear difficult because of the immunologic species barrier, but pig islets have been shown to function in primates for at least 6 mo with clinically incompatible immunosuppression. Therefore, a bioartificial pancreas made of encapsulated pig islets may resolve issues associated with islet allotransplantation. Although several groups have shown that encapsulated pig islets are functional in small-animal models, less is known about the use of bioartificial pancreases in large-animal models. In this review, we summarize current knowledge of encapsulated pig islets, to determine obstacles to implantation in humans and possible solutions to overcome these obstacles.


Assuntos
Órgãos Bioartificiais , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Ilhotas Pancreáticas/metabolismo , Animais , Composição de Medicamentos , Terapia de Imunossupressão , Imunossupressores/farmacologia , Insulina/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Pâncreas/patologia , Ratos , Suínos , Fatores de Tempo , Transplante Heterólogo , Transplante Homólogo
20.
Biomaterials ; 32(34): 8880-91, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21872925

RESUMO

Adipose tissue was only recently considered as a potential source of mesenchymal stem cells (MSCs) for bone tissue engineering. To improve the osteogenicity of acellular bone allografts, adipose MSCs (AMSCs) and bone marrow MSCs (BM-MSCs) at nondifferentiated and osteogenic-differentiated stages were investigated in vitro and in vivo. In vitro experiments demonstrated a superiority of AMSCs for proliferation (6.1±2.3 days vs. 9.0±1.9 days between each passage for BM-MSCs, respectively, P<0.001). A significantly higher T-cell depletion (revealed by mixed lymphocyte reaction, [MLR]) was found for AMSCs (vs. BM-MSCs) at both non- and differentiated stages. Although nondifferentiated AMSCs secreted a higher amount of vascular endothelial growth factor [VEGF] in vitro (between 24 and 72 h of incubation at 0.1-21% O(2)) than BM-MSCs (P<0.001), the osteogenic differentiation induced a significantly higher VEGF release by BM-MSCs at each condition (P<0.001). After implantation in the paraspinal muscles of nude rats, a significantly higher angiogenesis (histomorphometry for vessel development (P<0.005) and VEGF expression (P<0.001)) and osteogenesis (as revealed by osteocalcin expression (P<0.001) and micro-CT imagery for newly formed bone tissue (P<0.05)) were found for osteogenic-differentiated AMSCs in comparison to BM-MSCs after 30 days of implantation. Osteogenic-differentiated AMSCs are the best candidate to improve the angio-/osteogenicity of decellularized bone allografts.


Assuntos
Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Transplante Ósseo , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Adipócitos/citologia , Adipócitos/imunologia , Adipócitos/metabolismo , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Transplante Ósseo/métodos , Diferenciação Celular , Células Cultivadas , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica , Osteocalcina/metabolismo , Osteogênese , Ratos , Ratos Nus , Suínos , Transplante Homólogo , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...