Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Biomaterials ; 32(19): 4376-84, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21458856

RESUMO

Widely used in different biomedical applications, polyelectrolyte multilayers provide inter alia an attractive way for manufacturing of bio-functionalized, stimuli responsive surface coatings to control cellular behavior. In this study a novel polyelectrolyte-based platform for the engineering and controllable detachment of human mesenchymal stem cell (MSC) sheets is presented. Thin films obtained by layer-by-layer deposition of cationic poly(allylamine hydrochloride) (PAH) and anionic poly(styrene sulfonate) (PSS) polyelectrolytes on conductive indium tin oxide (ITO) electrodes allowed for the fast formation of viable sheets from human placenta-derived mesenchymal stem cells (PD-MSCs). Resulting stem cell sheets retained their phenotypical profile and mesodermal differentiation potency. Both electrochemically-induced local pH lowering and global decrease of the environmental pH allowed for a rapid detachment of intact stem cell sheets. The recovered stem cell sheets remained viable and maintained their capacity to differentiate toward the adipogenic and osteogenic lineages. This novel polyelectrolyte multilayer based platform represents a promising, novel approach for the engineering of human stem cell sheets desired for future clinical applications.


Assuntos
Concentração de Íons de Hidrogênio , Células-Tronco Mesenquimais/citologia , Placenta/citologia , Engenharia Tecidual/métodos , Materiais Biocompatíveis , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células Cultivadas , Técnicas Eletroquímicas , Feminino , Humanos , Microscopia de Força Atômica , Gravidez
2.
Cell Transplant ; 20(8): 1241-57, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21176408

RESUMO

While therapeutic cell transplantations using progenitor cells are increasingly evolving towards phase I and II clinical trials and chemically defined cell culture is established, standardization in biobanking is still in the stage of infancy. In this study, the EU FP6-funded CRYSTAL (CRYo-banking of Stem cells for human Therapeutic AppLication) consortium aimed to validate novel Standard Operating Procedures (SOPs) to perform and validate xeno-free and chemically defined cryopreservation of human progenitor cells and to reduce the amount of the potentially toxic cryoprotectant additive (CPA) dimethyl sulfoxide (DMSO). To achieve this goal, three human adult progenitor and stem cell populations-umbilical cord blood (UCB)-derived erythroid cells (UCB-ECs), UCB-derived endothelial colony forming cells (UCB-ECFCs), and adipose tissue (AT)-derived mesenchymal stromal cells (AT-MSCs)-were cryopreserved in chemically defined medium supplemented with 10% or 5% DMSO. Cell recovery, cell repopulation, and functionality were evaluated postthaw in comparison to cryopreservation in standard fetal bovine serum (FBS)-containing freezing medium. Even with a reduction of the DMSO CPA to 5%, postthaw cell count and viability assays indicated no overall significant difference versus standard cryomedium. Additionally, to compare cellular morphology/membrane integrity and ice crystal formation during cryopreservation, multiphoton laser-scanning cryomicroscopy (cryo-MPLSM) and scanning electron microscopy (SEM) were used. Neither cryo-MPLSM nor SEM indicated differences in membrane integrity for the tested cell populations under various conditions. Moreover, no influence was observed on functional properties of the cells following cryopreservation in chemically defined freezing medium, except for UCB-ECs, which showed a significantly reduced differentiation capacity after cryopreservation in chemically defined medium supplemented with 5% DMSO. In summary, these results demonstrate the feasibility and robustness of standardized xeno-free cryopreservation of different human progenitor cells and encourage their use even more in the field of tissue-engineering and regenerative medicine.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/efeitos dos fármacos , Criopreservação/métodos , Crioprotetores/farmacologia , Tecido Adiposo/citologia , Adulto , Animais , Bovinos , Separação Celular , Forma Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Microscopia Crioeletrônica , Meios de Cultura Livres de Soro , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/ultraestrutura , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Células Eritroides/ultraestrutura , Sangue Fetal/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/ultraestrutura , Microscopia de Fluorescência por Excitação Multifotônica
3.
Genes Cells ; 15(7): 671-87, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20497237

RESUMO

Establishment of fetal bovine serum (FBS)-free cell culture conditions is essential for transplantation therapies. Blood-derived endothelial colony-forming cells (ECFCs) are potential candidates for regenerative medicine applications. ECFCs were isolated from term umbilical cord blood units and characterized by flow cytometry, capillary formation and responsiveness to cytokines. ECFCs were expanded under standard, FBS-containing endothelial medium, or transferred to chemically defined endothelial media without FBS. Microarray expression profiling was applied to compare the transcriptome profiles in FBS-containing versus FBS-free culture. ECFC outgrowth in standard medium was successful in 92% of cord blood units. The karyotype of expanded ECFCs remained normal. Without FBS, ECFC initiation and expansion failed. Modest proliferation, changes in cell morphology and organization and cell death have been observed after passaging. Gene ontology analysis revealed a broad down-regulation of genes involved in cell cycle progression and up-regulation of genes involved in stress response and apoptosis. Interestingly, genes participating in lipid biosynthesis were markedly up-regulated. Detection of several endothelial cell-specific marker genes showed the maintenance of the endothelial cell characteristics during serum-free culture. Although ECFCs maintain their endothelial characteristics during serum-free culturing, they could not be expanded. Additional supply of FBS-free media with lipid concentrates might increase the ECFC survival.


Assuntos
Técnicas de Cultura de Células/métodos , Células Endoteliais/citologia , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Transplante de Células-Tronco Hematopoéticas , Animais , Bovinos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos
4.
Am J Obstet Gynecol ; 202(1): 85.e1-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20096254

RESUMO

OBJECTIVE: This study was undertaken to test injectable surgical sealants that are biocompatible with fetal membranes and that are to be used eventually for the closure of iatrogenic membrane defects. STUDY DESIGN: Dermabond (Ethicon Inc, Norderstedt, Germany), Histoacryl (B. Braun GmbH, Tuttlingen, Germany), and Tissucol (Baxter AG, Volketwil, Switzerland) fibrin glue, and 3 types of in situ forming poly(ethylene glycol)-based polymer hydrogels were tested for acute toxicity on direct contact with fetal membranes for 24 hours. For the determination of elution toxicity, extracts of sealants were incubated on amnion cell cultures for 72 hours. Bonding and toxicity was assessed through morphologic and/or biochemical analysis. RESULTS: Extracts of all adhesives were nontoxic for cultured cells. However, only Tissucol and 1 type of poly(ethylene glycol)-based hydrogel, which is a mussel-mimetic tissue adhesive, showed efficient, nondisruptive, nontoxic bonding to fetal membranes. Mussel-mimetic tissue adhesive that was applied over membrane defects that were created with a 3.5-mm trocar accomplished leak-proof closure that withstood membrane stretch in an in vitro model. CONCLUSION: A synthetic hydrogel-type tissue adhesive that merits further evaluation in vivo emerged as a potential sealing modality for iatrogenic membrane defects.


Assuntos
Âmnio/efeitos dos fármacos , Âmnio/cirurgia , Cianoacrilatos/farmacocinética , Adesivo Tecidual de Fibrina/farmacologia , Hidrogéis/uso terapêutico , Polietilenoglicóis/farmacologia , Adesivos Teciduais/farmacologia , Âmnio/citologia , Cianoacrilatos/administração & dosagem , Cianoacrilatos/farmacologia , Embucrilato/administração & dosagem , Embucrilato/farmacologia , Feminino , Ruptura Prematura de Membranas Fetais , Fetoscopia , Adesivo Tecidual de Fibrina/administração & dosagem , Humanos , Hidrogéis/administração & dosagem , Técnicas In Vitro , Teste de Materiais , Polietilenoglicóis/administração & dosagem , Gravidez , Adesivos Teciduais/administração & dosagem
5.
Am J Obstet Gynecol ; 202(2): 193.e1-193.e13, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20035913

RESUMO

OBJECTIVE: This study was undertaken to isolate and characterize multipotent mesenchymal stem cells from term human placenta (placenta-derived mesenchymal stem cells, PD-MSCs). STUDY DESIGN: Sequential enzymatic digestion was used to isolate PD-MSCs in which trypsin removes the trophoblast layer, followed by collagenase treatment of remaining placental tissue. Karyotype, phenotype, growth kinetics, and differentiability of PD-MSC isolates from collagenase digests were analyzed. RESULTS: PD-MSC isolation was successful in 14 of 17 cases. Karyotyping of PD-MSC isolates from deliveries with a male fetus revealed that these cells are of maternal origin. Flow cytometry and immunocytochemistry confirmed the mesenchymal stem cell phenotype. Proliferation rates of PD-MSCs remained constantly high up to passage 20. These cells could be differentiated toward mesodermal lineage in vitro up to passage 20. Nonconfluent culture was critical to maintain the MSC stemness during long-term culture. CONCLUSION: Term placenta constitutes a rich, very reliable source of maternal mesenchymal stem cells that remain differentiable, even at high passage numbers.


Assuntos
Separação Celular/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Multipotentes/citologia , Placenta/citologia , Antígeno AC133 , Adulto , Antígenos CD/análise , Diferenciação Celular , Feminino , Glicoproteínas/análise , Humanos , Recém-Nascido , Masculino , Peptídeos/análise , Gravidez
6.
Anesth Analg ; 109(4): 1117-26, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19762739

RESUMO

BACKGROUND: Endothelial progenitor cells play a pivotal role in tissue repair, and thus are used for cell replacement therapies in "regenerative medicine." We tested whether the anesthetic sevoflurane would modulate growth or mobilization of these angiogenic cells. METHODS: In an in vitro model, mononuclear cells isolated from peripheral blood of healthy donors were preconditioned with sevoflurane (3 times 30 min at 2 vol% interspersed by 30 min of air). Colony-forming units were determined after 9 days in culture and compared with time-matched untreated control. Using magnetic cell sorting, CD133+/CD34+ endothelial progenitors were enriched from human umbilical cord blood, and vascular endothelial growth factor (VEGF), VEGFR2 (KDR), granulocyte colony-stimulating factor (G-CSF), STAT3, c-kit, and CXCR4 expressions were determined in sevoflurane-treated and untreated cells by real-time reverse transcriptase polymerase chain reaction. In a volunteer study with crossover design, we tested whether sevoflurane inhalation (<1 vol% end-tidal concentration) would mobilize endothelial progenitor cells from the bone marrow niche into the circulation using flow cytometry of peripheral blood samples. VEGF and G-CSF plasma levels were also measured. RESULTS: In vitro sevoflurane exposure of mononuclear cells enhanced colony-forming capacity and increased VEGF mRNA levels in CD133+/CD34+ cord blood cells (P = 0.017). Sevoflurane inhalation in healthy volunteers did not alter the number of CD133+/CD34+ or KDR+/CD34+ endothelial progenitors in the circulation, but increased the number of colony-forming units (P = 0.034), whereas VEGF and G-CSF plasma levels remained unchanged. CONCLUSIONS: Sevoflurane preconditioning promotes growth and proliferation of stem cell-like human endothelial progenitors. Hence, it may be used to promote perioperative vascular healing and to support cell replacement therapies.


Assuntos
Anestésicos Inalatórios/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Éteres Metílicos/farmacologia , Células-Tronco/efeitos dos fármacos , Antígeno AC133 , Adulto , Anestésicos Inalatórios/administração & dosagem , Antígenos CD/análise , Antígenos CD34/análise , Separação Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Estudos Cross-Over , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Feminino , Sangue Fetal/citologia , Citometria de Fluxo , Glicoproteínas/análise , Fator Estimulador de Colônias de Granulócitos/sangue , Humanos , Masculino , Éteres Metílicos/administração & dosagem , Pessoa de Meia-Idade , Peptídeos/análise , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA Mensageiro/metabolismo , Receptores CXCR4/metabolismo , Fator de Transcrição STAT3/metabolismo , Sevoflurano , Células-Tronco/imunologia , Células-Tronco/metabolismo , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
7.
Tissue Eng Part A ; 15(10): 2977-90, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19320572

RESUMO

Polyelectrolyte multilayer coatings have emerged as substrates to control cellular behavior, but interactions with human multipotent mesenchymal stromal cells (MSCs) have not been studied. We looked at layer-by-layer coatings of cationic poly-L-lysine (PLL) and anionic hyaluronic acid (HA) as substrates for MSCs of placenta and adipose tissue. This system allows for modulation of thickness (number of deposition cycles), stiffness (chemical cross-linking of bulk layer), and adhesiveness (fibronectin (FN) interface). Native, as-built PLL/HA multilayer coatings were poorly adhesive for MSCs despite spectroscopy-confirmed high surface density of pre-adsorbed FN. Stratification of cross-linked PLL/HA multilayers of different stiffnesses revealed that multilayers modified with a high cross-linking regimen became efficient substrates for MSC adhesion and proliferation. MSCs on cross-linked multilayers grew to confluence. Using comparative confocal microscopy analysis of PLL/HA multilayers with physically adsorbed versus chemically coupled FN, we demonstrated that cross-linking strongly influenced FN surface distribution, leading to denser presentation of adhesion sites for cells. The covalent affixation of FN promoted focal adhesion formation and was critical to maintaining densely grown MSC cultures over weeks for their differentiation. Multilayer-bound MSCs were capable of differentiating into osteocytes and chondrocytes upon culture with induction factors. Together, cross-linked, FN-terminated PLL/HA multilayers provide a versatile platform for studies of human MSCs for biotechnological or therapeutic applications.


Assuntos
Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Polímeros/química , Engenharia Tecidual/métodos , Proliferação de Células , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Microscopia Confocal , Gravidez
8.
Cell Transplant ; 17(8): 955-68, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19069637

RESUMO

Emerging evidence suggests human amnion tissue as a valuable source of two distinct types of pluripotent cells, amnion epithelial cells (hAECs) and mesenchymal stromal cells (hAMSCs), for applications in cell replacement therapy. For some approaches, it may be necessary to culture and differentiate these cells before they can be transplanted. No systematic attempt has been yet made to determine the quantity and quality of amnion cells after isolation and culture. We looked at amnion cell isolates from 27 term placentas. Following our optimized protocol, primary yields were 6.3 x 10(6) hAECs and 1.7 x 10(6) hAMSCs per gram amnion. All 27 cases gave vital cultures of hAMSCs, while one third of cases (9 of 27) failed to give adherent cultures of hAECs. Primary cultures contained significantly more proliferating than apoptotic cells (hAECs: 16.4% vs. 4.0%; hAMSCs: 9.5% vs. 2.4%). Neither hAECs nor hAMSCs were clonogenic. They showed slow proliferation that almost stopped beyond passage 5. Microscopic follow-up revealed that hAEC morphology gradually changed towards mesenchymal phenotype over several passages. Flow cytometric characterization of primary cultures showed expression of mesenchymal progenitor markers CD73, CD90, CD105, and CD166, as well as the embryonic stem cell markers SSEA-3 and -4 on both amnion cell types. These profiles were grossly maintained in secondary cultures. Reverse transcriptase-PCR analysis exhibited transcripts of Oct-3/4 and stem cell factor in primary and secondary cultures of all cases, but no telomerase reverse transcriptase. Immunocytochemistry confirmed translation into Oct-3/4 protein in part of hAEC cultures, but not in hAMSCs. Further, both amnion cell types stained for CD90 and SSEA-4. Osteogenic induction studies with amnion cells from four cases showed significantly stronger differentiation of hAECs than hAMSCs; this capacity to differentiate greatly varied between cases. In conclusion, hAECs and hAMSCs in culture exhibit and maintain a similar marker profile of mesenchymal progenitors. hAECs were found as a less reliable source than hAMSCs and altered morphology during subculture.


Assuntos
Âmnio/transplante , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Epiteliais/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Células Estromais/transplante , Âmnio/citologia , Âmnio/metabolismo , Antígenos de Superfície/análise , Antígenos de Superfície/metabolismo , Biomarcadores , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Proliferação de Células , Forma Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Fator 3 de Transcrição de Octâmero/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Células-Tronco/genética , Células Estromais/citologia , Células Estromais/metabolismo
9.
Swiss Med Wkly ; 138(41-42): 596-601, 2008 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-18941945

RESUMO

Preterm premature rupture of the foetal membranes (früher vorzeitiger Blasensprung) has remained a devastating complication of pregnancy with very high risk of pregnancy loss. Several methods of sealing spontaneously ruptured membranes to stop amniotic fluid leakage and prolong pregnancy have been tested, but no one of them has achieved a clinical breakthrough. Also, needle and foetoscopic puncture of membranes for diagnostic or surgical interventions in the amniotic cavity carry a significant risk of persistent membrane leakage and subsequent rupture - thus limiting the developing field of intrauterine foetal surgery. Efforts are concentrated on taking action before rupture rather than reacting after rupture: one avenue of research concerns prophylactic plugging of foetoscopic access sites in foetal membranes at the time of intervention, thus inhibiting leakage and rupture. Foetal membrane injuries, spontaneous or iatrogenic, constitute extreme challenges to repair: thinness of foetal membrane tissue, difficult localisation and accessibility of the rupture site, the need for injectable sealants, wet gluing conditions and poor wound healing in this tissue all complicate repair. The goal is to achieve immediate and at the same time long-lasting closure of the membrane leak. Here we review approaches to closure of foetal membrane defects with liquid sealants or solid biomaterial scaffolds, with the focus on prophylactic plugging of foetoscopic access sites.


Assuntos
Ruptura Prematura de Membranas Fetais/terapia , Engenharia Tecidual/métodos , Feminino , Humanos , Hidrogéis/uso terapêutico , Gravidez , Resultado da Gravidez , Adesivos Teciduais/uso terapêutico , Alicerces Teciduais
10.
Biomaterials ; 29(26): 3531-8, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18541296

RESUMO

Growth factors are often used in tissue regeneration to stimulate vascularisation of polymeric scaffolds, with vascular endothelial growth factor (VEGF) having been extensively studied for short-term vessel ingrowth. We have therefore evaluated the effect of different concentrations of VEGF on the vascularisation of a porous scaffold in the short-, intermediate- and long-term, by delivering 15, 150 and 1500ng VEGF/day to polyurethane scaffolds by osmotic pumps for up to 6 weeks. An increased vascularisation months after termination of VEGF delivery was only achieved with 150ng/day (46%, p<0.05). This dosage consistently showed elevated levels of vascularisation (144, 125, 160 and 60% above PBS controls at 10, 20, 30 and 42 days, respectively, p<0.05), whilst the vessels induced by the highest dosage, though initially maximally elevated (265 and 270% at 10 and 20 days, p<0.05) tended to regress after 20 days of VEGF delivery. Pericyte coverage was decreased at 20 days for the highest dosage (30%, p<0.05). Lectin perfusion demonstrated that vessels within the scaffold were connected to the host vasculature at all time points and perfusion was substantially raised by VEGF delivery at day 20. These results suggest concentration of VEGF plays a critical role in the nature and persistence of vasculature formed in a tissue regenerative scaffold.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Alicerces Teciduais , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Relação Dose-Resposta a Droga , Engenharia Tecidual , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Biomaterials ; 29(11): 1720-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18155761

RESUMO

A major challenge for therapeutic delivery of angiogenic agents such as vascular endothelial growth factor (VEGF) is to achieve sustained, low dose signaling leading to durable neovessel formation. To this end, we recently created a variant of VEGF(121), TG-VEGF(121) that directly binds to fibrin and gets released locally in proteolysis-triggered manner. Here we combined noninvasive biophotonic monitoring of VEGF receptor 2 gene activation in transgenic VEGFR2-luc mice and histomorphometry to compare endothelial activation and long-term neovascularization by actively released TG-VEGF(121)versus passively released, diffusible wild-type VEGF(121) in subcutaneous fibrin implants. Monitoring in real-time over 3 weeks of luciferase signal driven by the VEGFR2 promoter revealed endothelial activation in skin exposed to wild-type VEGF(121), but no detectable elevation over fibrin alone by TG-VEGF(121). Histology at 3 weeks, however, demonstrated that TG-VEGF(121) promoted vessel growth significantly more effectively and reliably than wild-type VEGF(121). The majority of vessels surviving to 3 weeks contained stabilizing smooth muscle cells. Yet, by 6 weeks, no extra vessels induced by exogenous VEGF were left. In conclusion, release of fibrin-conjugated variant TG-VEGF(121) elicited lower VEGFR2-luc activation than wild-type VEGF(121) yet significantly more vascularization. In the absence of true physiological demand, even stabilized vessels are ultimately regressed.


Assuntos
Fibrina/metabolismo , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Ativação Enzimática , Fibrina/química , Camundongos , Modelos Animais , Próteses e Implantes , Fatores de Tempo
12.
Obstet Gynecol ; 110(5): 1121-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17978128

RESUMO

OBJECTIVE: To explore a surgical plug formed from decellularized term human amnion membrane for fetoscopic closure of iatrogenic defects in fetal membranes in a rabbit model. METHODS: The study was performed in eight rabbit does. Punctures were created at midgestational day 23 by 14-gauge needle fetoscopy on surgically exposed rabbit amniotic sacs. The entry sites were fetoscopically plugged either with decellularized term human amnion membrane (n=10) or previously successful commercial collagen matrix foil (n=10), followed by their primary fixation with fibrin glue and myometrial suturing. Seven punctured sacs without any plugging and 31 sacs without any manipulation served as two reference groups. Amniotic integrity and fetal parameters were assessed at gestational day 30. RESULTS: We established a facile method to prepare sheets of decellularized term human amnion membrane and verified its nontoxicity and cell compatibility in vitro. Decellularized term human amnion membrane sheets could be delivered precisely and controlled by fetoscopy as compact plugs into amniotic defects. The surgical handling characteristics of decellularized term human amnion membrane were better than the commercial collagen matrix foil. Treatment with human decellularized term human amnion membrane was comparable to treatment with the collagen matrix with regard to efficiency in restoring amniotic integrity. Seventy-five percent and 71.4% of amniotic sacs treated with decellularized term human amnion membrane or the commercial collagen matrix foil, respectively, showed amniotic integrity, compared with 25% in the left-open study group. Histology at the 1 week experimental endpoint showed no evidence for inflammation or beginning of anatomic healing of grafted, decellularized term human amnion membrane. CONCLUSION: Fetoscopic delivery of plugs made of decellularized term human amnion membrane presents a potentially practical surgical method to restore amniotic integrity of punctured fetal membranes. LEVEL OF EVIDENCE: III.


Assuntos
Âmnio , Materiais Biocompatíveis/administração & dosagem , Membranas Extraembrionárias/lesões , Fetoscopia/efeitos adversos , Alicerces Teciduais , Adulto , Animais , Estudos de Casos e Controles , Feminino , Fetoscopia/métodos , Humanos , Doença Iatrogênica , Modelos Animais , Gravidez , Coelhos , Adesivos Teciduais , Cicatrização
13.
Biomaterials ; 28(26): 3856-66, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17568666

RESUMO

The molecular engineering of cell-instructive artificial extracellular matrices is a powerful means to control cell behavior and enable complex processes of tissue formation and regeneration. This work reports on a novel method to produce such smart biomaterials by recapitulating the crosslinking chemistry and the biomolecular characteristics of the biopolymer fibrin in a synthetic analog. We use activated coagulation transglutaminase factor XIIIa for site-specific coupling of cell adhesion ligands and engineered growth factor proteins to multiarm poly(ethylene glycol) macromers that simultaneously form proteolytically sensitive hydrogel networks in the same enzyme-catalyzed reaction. Growth factor proteins are quantitatively incorporated and released upon cell-derived proteolytic degradation of the gels. Primary stromal cells can invade and proteolytically remodel these networks both in an in vitro and in vivo setting. The synthetic ease and potential to engineer their physicochemical and bioactive characteristics makes these hybrid networks true alternatives for fibrin as provisional drug delivery platforms in tissue engineering.


Assuntos
Matriz Extracelular/química , Fibrina/química , Fibrina/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Engenharia Tecidual/métodos , Transglutaminases/química , Técnicas de Cultura de Células/métodos , Linhagem Celular , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos
14.
J Neurosci ; 27(19): 5127-38, 2007 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-17494698

RESUMO

Specialized postsynaptic structures known as dendritic spines are the primary sites of glutamatergic innervation at synapses of the CNS. Previous studies have shown that spines rapidly remodel their actin cytoskeleton to modify their shape and this has been associated with changes in synaptic physiology. However, the receptors and signaling intermediates that restructure the actin network in spines are only beginning to be identified. We reported previously that the EphA4 receptor tyrosine kinase regulates spine morphology. However, the signaling pathways downstream of EphA4 that induce spine retraction on ephrin ligand binding remain poorly understood. Here, we demonstrate that ephrin stimulation of EphA4 leads to the recruitment and activation of phospholipase Cgamma1 (PLCgamma1) in heterologous cells and in hippocampal slices. This interaction occurs through an Src homology 2 domain of PLCgamma1 and requires the EphA4 juxtamembrane tyrosines. In the brain, PLCgamma1 is found in multiple compartments of synaptosomes and is readily found in postsynaptic density fractions. Consistent with this, PLC activity is required for the maintenance of spine morphology and ephrin-induced spine retraction. Remarkably, EphA4 and PLC activity modulate the association of the actin depolymerizing/severing factor cofilin with the plasma membrane. Because cofilin has been implicated previously in the structural plasticity of spines, this signaling may enable cofilin to depolymerize actin filaments and restructure spines at sites of ephrin-EphA4 contact.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Espinhas Dendríticas/metabolismo , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Fosfolipase C gama/metabolismo , Receptor EphA4/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Células COS , Membrana Celular/química , Membrana Celular/metabolismo , Forma Celular/fisiologia , Células Cultivadas , Chlorocebus aethiops , Espinhas Dendríticas/ultraestrutura , Ativação Enzimática/fisiologia , Efrinas/metabolismo , Hipocampo/ultraestrutura , Camundongos , Plasticidade Neuronal/fisiologia , Técnicas de Cultura de Órgãos , Fosfolipase C gama/química , Fosforilação , Estrutura Terciária de Proteína/fisiologia , Receptor EphA4/química , Transdução de Sinais/fisiologia , Sinaptossomos/metabolismo , Sinaptossomos/ultraestrutura
15.
J Thorac Cardiovasc Surg ; 133(5): 1163-70, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17467424

RESUMO

OBJECTIVE: Intimal hyperplasia and surface thrombogenicity are major factors in the high failure rate of synthetic small-diameter bypass grafts. Vascular endothelial growth factor is a potent stimulus for endothelial growth, and its provision in a fibrin matrix coating at the luminal graft surface may hold a key to spontaneous graft endothelialization and improved graft patency. METHODS: Pigs underwent bilateral carotid artery interposition of expanded polytetrafluoroethylene grafts either impregnated with fibrin (n = 11)--engineered to locally release vascular endothelial growth factor121 (vascular endothelial growth factor-fibrin; n = 11)--or left uncoated (n = 12). Graft patency was assessed by quantitative carotid angiography followed by graft histomorphometry at the 1-month experimental end point. RESULTS: Patency rates were not significantly different between study groups. Grafts coated with fibrin or vascular endothelial growth factor-fibrin exhibited significantly increased angiographic narrowing at the proximal anastomosis (for both P < .05 vs uncoated) and no difference at the distal anastomosis and the grafts' middle. Histological analysis showed 80% to 90% endothelial coverage and buildup of intima throughout the lengths of all grafts. Examination of the grafts' midportion revealed significantly enlarged neointimal layers of smooth muscle actin-positive cells in grafts coated with vascular endothelial growth factor-fibrin (242 +/- 47 microm2/micron) and fibrin (177 +/- 41 microm2/micron), compared with uncoated grafts (131 +/- 39 microm2/micron) (for both P < .05 vs uncoated). This thickening could not be explained by enhanced inflammation or vessel wall angiogenesis, which were minimal at the experimental end point. CONCLUSIONS: Fibrin and vascular endothelial growth factor produced effects deleterious to graft healing, by increasing the narrowing at proximal anastomosis and neointimal growth beyond that seen in uncoated grafts. It may reflect direct activation by exogenous vascular endothelial growth factor of vascular smooth muscle cells.


Assuntos
Prótese Vascular , Artérias Carótidas/patologia , Materiais Revestidos Biocompatíveis , Fibrina , Politetrafluoretileno , Túnica Íntima/patologia , Fator A de Crescimento do Endotélio Vascular , Animais , Implante de Prótese Vascular , Capilares/patologia , Artérias Carótidas/cirurgia , Fibrina/farmacologia , Oclusão de Enxerto Vascular/patologia , Imuno-Histoquímica , Macrófagos/patologia , Sus scrofa , Fator A de Crescimento do Endotélio Vascular/farmacologia , Grau de Desobstrução Vascular
16.
Biochem Biophys Res Commun ; 350(2): 364-9, 2006 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-17010941

RESUMO

Cord blood is source of colony-forming progenitors to vascular endothelial cells for potential use in cell therapies. These cells-called blood late outgrowth endothelial cells (OECs)-have undergone endothelial differentiation, but appear to still possess functional properties different from mature endothelial cells. A large-scale comparative proteomics screen of cord blood OECs versus human vein endothelial cells (HUVECs) using two-dimensional gel electrophoresis and mass spectrometry identified specific expression of manganese superoxide dismutase (MnSOD), a key antioxidant enzyme expressed in the mitochondria, in OECs but not in HUVECs. Immunoblotting verified significant MnSOD levels in all OEC isolates tested and maintained throughout passaging. Endothelial function and cell survival/proliferation assays in the presence of high cytotoxic doses of the superoxide generator compound LY83583 showed OECs profoundly better protected against oxidative stress than HUVECs. Such cytoprotective levels of MnSOD cells could give therapeutic cell transplants a survival advantage in necrotic or ischemic conditions.


Assuntos
Células Endoteliais/enzimologia , Endotélio Vascular/citologia , Sangue Fetal/citologia , Estresse Oxidativo , Superóxido Dismutase/metabolismo , Capilares/crescimento & desenvolvimento , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Proteômica , Espécies Reativas de Oxigênio/metabolismo
17.
Proc Natl Acad Sci U S A ; 102(44): 15779-84, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16249343

RESUMO

Cell organization is largely orchestrated by extracellular gradients of morphogenetic proteins. VEGF, an essential factor for capillary formation, is stored in the extracellular matrix, but the mechanisms by which it and other matrix-bound morphogens are mobilized to form spatial gradients are poorly understood. Here, we suggest an efficient mechanism for morphogen gradient generation by subtle biophysical forces in an in vitro model of capillary morphogenesis. Using a fibrin-bound VEGF variant that is released proteolytically to mimic the in vivo situation, we report that low levels of interstitial flow act synergistically with VEGF to drive endothelial organization, whereas each stimulus alone has very little effect. To help account for this synergy, we show how these slow flows can bias the distribution of cell-secreted proteases, which leads, interestingly, to the creation of an increasing VEGF gradient relative to the cell and skewed in the direction of flow. In contrast, diffusion alone can only account for symmetric, decreasing autocrine gradients. Indeed, branching of capillary structures was biased in the direction of flow only with the combination of VEGF and flow. This work thus demonstrates a general mechanism of morphogen gradient generation and amplification by small ubiquitous mechanical forces that are known to exist in vivo.


Assuntos
Capilares/crescimento & desenvolvimento , Endotélio Vascular/citologia , Líquido Extracelular/fisiologia , Morfogênese , Fator A de Crescimento do Endotélio Vascular/fisiologia , Células Cultivadas , Difusão , Matriz Extracelular/fisiologia , Fibrina/metabolismo , Humanos , Metaloproteinases da Matriz/metabolismo , Reologia , Estresse Mecânico
18.
J Biol Chem ; 280(23): 22445-53, 2005 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15781448

RESUMO

The vessel-stabilizing effect of angiopoietin-1 (Ang1)/Tie2 receptor signaling is a potential target for pro-angiogenic therapies as well as anti-angiogenic inhibition of tumor growth. We explored the endothelial and vascular specific activities of the Ang1 monomer, i.e. dissociated from its state as an oligomer. A truncated monomeric Ang1 variant (i.e. DeltaAng1) containing the isolated fibrinogen-like receptor-binding domain of Ang1 was created and recombinantly produced in insect cells. DeltaAng1 ligated the Tie2 receptor without triggering its phosphorylation. Moreover, monomeric DeltaAng1 was observed to bind alpha(5)beta(1) integrin with similar affinity compared with Tie2. Unexpectedly, in vitro treatment of endothelial cells with DeltaAng1 showed some of the known effects of full-length Ang1, including inhibition of basal endothelial cell permeability and stimulation of cell adhesion as well as activation of MAPKs. Local treatment of the microvasculature of the developing chicken chorioallantoic membrane with the DeltaAng1 protein led to profound reduction of the mean vascular length density, thinning of vessels, and reduction of the number of vessel branching points. Similar effects were observed in side-by-side experiments with the recombinant full-length Ang1 protein. These effects of simplification of the vessel branching pattern were confirmed through local gene transfer with lentiviral particles encoding DeltaAng1 or full-length Ang1. Together, our findings suggest a potential use for exogenous Ang1 in reducing rather than increasing vascular density. Furthermore, we show that the isolated receptor-binding domain of Ang1 is capable of mediating some effects of full-length Ang1 independently of Tie2 phosphorylation, possibly through integrin ligation.


Assuntos
Angiopoietina-1/genética , Angiopoietina-1/fisiologia , Endotélio Vascular/metabolismo , Técnicas de Transferência de Genes , Animais , Aorta/citologia , Aorta/metabolismo , Baculoviridae/metabolismo , Biotinilação , Adesão Celular , Linhagem Celular , Células Cultivadas , Embrião de Galinha , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Endotélio Vascular/citologia , Ativação Enzimática , Fibrina/química , Fibrinogênio/química , Vetores Genéticos , Humanos , Insetos , Integrinas/metabolismo , Lentivirus/genética , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Músculo Liso/citologia , Neovascularização Patológica , Permeabilidade , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Receptor TIE-2/metabolismo , Proteínas Recombinantes/química , Sefarose/química , Transdução de Sinais
19.
J Control Release ; 101(1-3): 93-109, 2005 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-15588897

RESUMO

A number of vascular therapies could benefit from advanced methods for presentation of angiogenic growth factors, including growth of endothelium on small caliber vascular grafts and revascularization of ischemic tissue through induction of collateral vessels and microvessels. To explore methods to optimize the presentation and release of angiogenic factors in such applications in device integration and tissue repair, we studied three variant forms of vascular endothelial growth factor 121 (VEGF121), each with differential susceptibility to local cellular proteolytic activity, formulated within fibrin matrices. (1) The prototypic variant alpha2PI(1-8)-VEGF121 remains immobilized in fibrin matrices until its liberation by cell-associated enzymes, such as plasmin, that degrade the fibrin network [slow, cell-demanded release; J. Control. Release 72 (2001) 101-113]; the alpha2PI(1-8) domain serves as a site for covalent attachment to fibrin during coagulation. (2) We created a new VEGF variant, alpha2PI(1-8)-Pla-VEGF121 that couples to fibrin via a plasmin-sensitive sequence (Pla). Cleavage of this target site by plasmin enables direct release of alpha2PI(1-8)-Pla-VEGF121 from bulk matrix degradation (accelerated, cell-demanded release). (3) Native VEGF121 (burst, passive release) was considered as a reference. VEGF release profiles were determined experimentally as well as mathematically, alpha2PI(1-8)-Pla-VEGF121 being released ca. fourfold more quickly than alpha2PI(1-8)-VEGF121, both being retained compared to native VEGF121; the differences in release could be accounted for based on knowledge of the plasmin sensitivity of the bound growth factor and the structure of the fibrin network. The bound factors were competent in inducing endothelial cell proliferation, the matrix-bound forms being more effective than native VEGF121; as well as competent in inducing endothelial progenitor cell maturation into endothelial cells. These matrix-bound variants of VEGF121 may be particularly useful where retention in locally applied surgical sites is desired, such as prevention of washout from vascular graft coatings and slowing loss from tissue ingrowth matrices used in local tissue revascularization and repair.


Assuntos
Células Endoteliais/fisiologia , Fibrina/metabolismo , Células-Tronco/fisiologia , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Proliferação de Células , Sistemas de Liberação de Medicamentos , Fibrinolisina/farmacologia , Humanos , Matemática , Modelos Biológicos , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Biotechnol Bioeng ; 89(1): 9-17, 2005 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-15580576

RESUMO

The dramatically increasing prevalence of multi-drug-resistant human pathogenic bacteria and related mortality requires two key actions: (i) decisive initiatives for the detection of novel antibiotics and (ii) a global ban for use of antibiotics as growth promotants in stock farming. Both key actions entail technology for precise, high-sensitive detection of antibiotic substances either to detect and validate novel anti-infective structures or to enforce the non-use of clinically relevant antibiotics. We have engineered prokaryotic antibiotic response regulators into a molecular biosensor configuration able to detect tetracycline, streptogramin, and macrolide antibiotics in spiked liquids including milk and serum at ng/mL concentrations and up to 2 orders of magnitude below current Swiss and EC threshold values. This broad-spectrum, class-specific, biosensor-based assay has been optimized for use in a storable ready-to-use and high-throughput-compatible ELISA-type format. At the center of the assay is an antibiotic sensor protein whose interaction with specific DNA fragments is responsive to a particular class of antibiotics. Binding of biosensor protein to the cognate DNA chemically linked to a solid surface is converted into an immuno-based colorimetric readout correlating with specific antibiotics concentrations.


Assuntos
Antibacterianos/análise , Proteínas de Bactérias , Técnicas Biossensoriais , Peptídeos , Proteínas/análise , Animais , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/patogenicidade , Western Blotting , Células CHO , Colorimetria , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Farmacorresistência Bacteriana Múltipla , Quimioterapia Combinada , Ensaio de Imunoadsorção Enzimática , Genes Reporter , Humanos , Proteínas/isolamento & purificação , Proteínas/metabolismo , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA