Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 302(10): H2031-42, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22427522

RESUMO

To bridge the gap between two-dimensional cell culture and tissue, various three-dimensional (3-D) cell culture approaches have been developed for the investigation of cardiac myocytes (CMs) and cardiac fibroblasts (CFs). However, several limitations still exist. This study was designed to develop a cardiac 3-D culture model with a scaffold-free technology that can easily and inexpensively generate large numbers of microtissues with cellular distribution and functional behavior similar to cardiac tissue. Using micromolded nonadhesive agarose hydrogels containing 822 concave recesses (800 µm deep × 400 µm wide), we demonstrated that neonatal rat ventricular CMs and CFs alone or in combination self-assembled into viable (Live/Dead stain) spherical-shaped microtissues. Importantly, when seeded simultaneously or sequentially, CMs and CFs self-sorted to be interspersed, reminiscent of their myocardial distribution, as shown by cell type-specific CellTracker or antibody labeling. Microelectrode recordings and optical mapping revealed characteristic triangular action potentials (APs) with a resting membrane potential of -66 ± 7 mV (n = 4) in spontaneously contracting CM microtissues. Under pacing, optically mapped AP duration at 90% repolarization and conduction velocity were 100 ± 30 ms and 18.0 ± 1.9 cm/s, respectively (n = 5 each). The presence of CFs led to a twofold AP prolongation in heterogenous microtissues (CM-to-CF ratio of 1:1). Importantly, Ba(2+)-sensitive inward rectifier K(+) currents and Ca(2+)-handling proteins, including sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a, were detected in CM-containing microtissues. Furthermore, cell type-specific adenoviral gene transfer was achieved, with no impact on microtissue formation or cell viability. In conclusion, we developed a novel scaffold-free cardiac 3-D culture model with several advancements for the investigation of CM and CF function and cross-regulation.


Assuntos
Comunicação Celular/fisiologia , Fibroblastos/citologia , Modelos Animais , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Potenciais de Ação/fisiologia , Animais , Células Cultivadas , Estimulação Elétrica , Fibroblastos/fisiologia , Hidrogéis , Potenciais da Membrana/fisiologia , Microeletrodos , Miócitos Cardíacos/fisiologia , Ratos , Ratos Sprague-Dawley
2.
Nat Med ; 2(11): 1263-7, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8898758

RESUMO

Lamellar ichthyosis (LI) is a disfiguring skin disease characterized by abnormal epidermal differentiation and defective cutaneous barrier function. LI has been associated with loss of keratinocyte transglutaminase 1 (TGase1), an enzyme believed necessary for normal formation of the cornified epidermal barrier. Using LI as a prototype for therapeutic cutaneous gene delivery, we have used the human skin/immunodeficient mouse xenograft model to correct the molecular, histologic and functional abnormalities of LI patient skin in vivo. We have used TGase1-deficient primary keratinocytes from LI patients combined with high-efficiency transfer of functional TGase1 to regenerate engineered human LI epidermis on immunodeficient mice. Engineered LI epidermis displayed normal TGase1 expression in vivo, unlike unengineered LI epidermis where TGase1 was absent. Epidermal architecture was also normalized by TGase1 restoration, as was expression of the epidermal differentiation marker filaggrin. Engineered LI skin demonstrated restoration of cutaneous barrier function measures to levels seen in epidermis regenerated by keratinocytes from patients with normal skin, indicating functional correction in vivo of the proposed primary pathophysiologic defect in LI. These results confirm a major role for TGase1 in epidermal differentiation and demonstrate a potential future approach to therapeutic gene delivery in human skin.


Assuntos
Técnicas de Transferência de Genes , Ictiose Lamelar/enzimologia , Queratinócitos/enzimologia , Transglutaminases/genética , Células 3T3 , Animais , Linhagem Celular , Células Cultivadas , Proteínas Filagrinas , Humanos , Ictiose Lamelar/patologia , Camundongos , Camundongos SCID , Pele/citologia , Transglutaminases/metabolismo
3.
J Cell Biol ; 143(5): 1317-28, 1998 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-9832559

RESUMO

Autocrine EGF-receptor (EGFR) ligands are normally made as membrane-anchored precursors that are proteolytically processed to yield mature, soluble peptides. To explore the function of the membrane-anchoring domain of EGF, we expressed artificial EGF genes either with or without this structure in human mammary epithelial cells (HMEC). These cells require activation of the EGFR for cell proliferation. We found that HMEC expressing high levels of membrane- anchored EGF grew at a maximal rate that was not increased by exogenous EGF, but could be inhibited by anti-EGFR antibodies. In contrast, when cells expressed EGF lacking the membrane-anchoring domain (sEGF), their proliferation rate, growth at clonal densities, and receptor substrate phosphorylation were not affected by anti-EGFR antibodies. The sEGF was found to be colocalized with the EGFR within small cytoplasmic vesicles. It thus appears that removal of the membrane-anchoring domain converts autocrine to intracrine signaling. Significantly, sEGF inhibited the organization of HMEC on Matrigel, suggesting that spatial restriction of EGF access to its receptor is necessary for organization. Our results indicate that an important role of the membrane-anchoring domain of EGFR ligands is to restrict the cellular compartments in which the receptor is activated.


Assuntos
Mama/citologia , Fator de Crescimento Epidérmico/química , Fator de Crescimento Epidérmico/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Sequência de Bases , Comunicação Celular , Linhagem Celular , Primers do DNA/genética , Fator de Crescimento Epidérmico/genética , Células Epiteliais/citologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/fisiologia , Feminino , Humanos , Ligantes , Membranas/metabolismo , Transdução de Sinais
4.
Science ; 237(4821): 1476-9, 1987 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-3629250

RESUMO

Retrovirus-mediated gene transfer was used to introduce a recombinant human growth hormone gene into cultured human keratinocytes. The transduced keratinocytes secreted biologically active growth hormone into the culture medium. When grafted as an epithelial sheet onto athymic mice, these cultured keratinocytes reconstituted an epidermis that was similar in appearance to that resulting from normal cells, but from which human growth hormone could be extracted. Transduced epidermal cells may prove to be a general vehicle for the delivery of gene products by means of grafting.


Assuntos
Epiderme/metabolismo , Regulação da Expressão Gênica , Hormônio do Crescimento/genética , DNA Recombinante/metabolismo , Humanos , Retroviridae/genética , Transcrição Gênica , Transdução Genética
5.
Neuron ; 32(2): 289-300, 2001 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-11683998

RESUMO

We have examined the roles of Hsc70 and auxilin in the uncoating of clathrin-coated vesicles (CCVs) during neuronal endocytosis. We identified two peptides that inhibit the ability of Hsc70 and auxilin to uncoat CCVs in vitro. When injected into nerve terminals, these peptides inhibited both synaptic transmission and CCV uncoating. Mutation of a conserved HPD motif within the J domain of auxilin prevented binding to Hsc70 in vitro and injecting this mutant protein inhibited CCV uncoating in vivo, demonstrating that the interaction of auxilin with Hsc70 is critical for CCV uncoating. These studies establish that auxilin and Hsc70 participate in synaptic vesicle recycling in neurons and that an interaction between these proteins is required for CCV uncoating.


Assuntos
Proteínas de Transporte/fisiologia , Vesículas Revestidas por Clatrina/metabolismo , Proteínas de Choque Térmico HSP70/fisiologia , Proteínas de Membrana/fisiologia , Terminações Pré-Sinápticas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Trifosfato de Adenosina/farmacologia , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/farmacologia , Bovinos , Grupo dos Citocromos c/química , Grupo dos Citocromos c/farmacologia , Endocitose , Proteínas de Choque Térmico HSC70 , Proteínas de Choque Térmico HSP70/farmacologia , Proteínas de Membrana/química , Proteínas de Membrana/farmacologia , Dados de Sequência Molecular , Mutagênese , Neurônios/ultraestrutura , Neurotransmissores/metabolismo , Fragmentos de Peptídeos/farmacologia , Transmissão Sináptica/efeitos dos fármacos
6.
J Clin Invest ; 98(12): 2894-902, 1996 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8981938

RESUMO

Based on preliminary but variable results with direct DNA transfer into wounds, we evaluated in vivo gene transfer by particle-mediated DNA delivery to rat skin to determine whether overexpression of TGF-beta1 at the site of skin incisions would result in a significant improvement in repair. Optimization of the method with viral promoter-luciferase reporter constructs indicated that expression of luciferase activity persisted up to 5 d and was promoter, pressure, and site dependent (ventral > dorsal). Using cytomegalovirus (CMV)-driven human alpha1-antitrypsin, transgene expression was immunolocalized within keratinocytes of the stratum granulosum at 24 h. We measured tensile strength of skin incisions at 11-21 d in both normal and diabetic rats transfected with TGF-beta1 expression vectors at surgery. Native murine TGF-beta1 under an SV40 promoter produced positive effects, while wound strengthening was more pronounced in diabetic animals using a CMV-driven construct. Transfection of rat skin with constitutively active, mutant porcine TGF-beta1 under the control of the CMV and Moloney murine leukemia virus promoters significantly increased tensile strength up to 80% for 14-21 d after surgery. Transfection 24 h before surgery was more effective. Particle-mediated gene delivery can be used to deliver viral promoter-cytokine expression constructs into rat skin in a safe, efficient, and reproducible fashion. The extent of wound repair, as evidenced by enhanced tensile strength, can be markedly improved in tissues transfected with TGF-beta1 expression constructs.


Assuntos
Pele/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Cicatrização/fisiologia , Animais , Biolística , Southern Blotting , DNA Complementar/genética , Diabetes Mellitus , Regulação da Expressão Gênica/genética , Técnicas de Transferência de Genes , Genes Reporter/genética , Células HeLa , Humanos , Imuno-Histoquímica , Queratinócitos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Ratos , Fator de Crescimento Transformador beta/genética , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/farmacologia
7.
J Neurosci ; 20(23): 8667-76, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11102472

RESUMO

Although clathrin assembly by adaptor proteins (APs) plays a major role in the recycling of synaptic vesicles, the molecular mechanism that allows APs to assemble clathrin is poorly understood. Here we demonstrate that AP180, like AP-2 and AP-3, binds to the N-terminal domain of clathrin. Sequence analysis reveals a motif, containing the sequence DLL, that exists in multiple copies in many clathrin APs. Progressive deletion of these motifs caused a gradual reduction in the ability of AP180 to assemble clathrin in vitro. Peptides from AP180 or AP-2 containing this motif also competitively inhibited clathrin assembly by either protein. Microinjection of these peptides into squid giant presynaptic terminals reversibly blocked synaptic transmission and inhibited synaptic vesicle endocytosis by preventing coated pit formation at the plasma membrane. These results indicate that the DLL motif confers clathrin assembly properties to AP180 and AP-2 and, perhaps, to other APs. We propose that APs promote clathrin assembly by cross-linking clathrin triskelia via multivalent interactions between repeated DLL motifs in the APs and complementary binding sites on the N-terminal domain of clathrin. These results reveal the structural basis for clathrin assembly and provide novel insights into the molecular mechanism of clathrin-mediated synaptic vesicle endocytosis.


Assuntos
Motivos de Aminoácidos/genética , Clatrina/metabolismo , Endocitose/fisiologia , Proteínas Monoméricas de Montagem de Clatrina , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/metabolismo , Vesículas Sinápticas/metabolismo , Subunidades alfa do Complexo de Proteínas Adaptadoras , Proteínas Adaptadoras de Transporte Vesicular , Animais , Sítios de Ligação/genética , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/genética , Bovinos , Células Cultivadas , Clatrina/química , Clatrina/genética , Invaginações Revestidas da Membrana Celular/efeitos dos fármacos , Invaginações Revestidas da Membrana Celular/metabolismo , Decapodiformes , Endocitose/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microinjeções , Proteínas do Tecido Nervoso/genética , Peptídeos/administração & dosagem , Peptídeos/química , Peptídeos/genética , Fosfoproteínas/genética , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos , Ressonância de Plasmônio de Superfície , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética
8.
Cloning Stem Cells ; 7(4): 226-31, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16390258

RESUMO

Islet-like cell clusters (ILCCs) were derived from murine embryonic stem cells using a slightly modified version of the protocol originally described by Lumelsky et al. in 2001. Analysis with enzyme-linked immunosorbent assays (ELISAs) that distinguish human from murine insulin demonstrated that insulin released from these ILCCs, upon initial in vitro glucose challenge, was of non-murine origin and in fact corresponded to the species of insulin, human or bovine, that had been added to the culture media used to derive ILCCs. This finding convincingly supports the hypothesis that ILCCs are not synthesizing insulin de novo, but rather simply regurgitating insulin taken up during tissue culture. In further experiments, ILCCs were derived in media in which insulin had been replaced by IGF-I with which it shares a common signaling pathway. These ILCCs failed to release any detectable insulin. In contrast, ILCCs produced by various protocols stained positive (dithizone and immunoselective antibodies) for intracellular insulin and, in some cases, C-peptide. Despite the presence of at least some level of de novo, synthesized insulin in ILCCs, the majority of insulin released by ILCCs was sequestered from the exogenous medium.


Assuntos
Embrião de Mamíferos/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Células-Tronco/metabolismo , Animais , Bovinos , Diferenciação Celular , Células Cultivadas , Reações Cruzadas , Embrião de Mamíferos/citologia , Ensaio de Imunoadsorção Enzimática , Transportador de Glucose Tipo 2/metabolismo , Humanos , Insulina/imunologia , Secreção de Insulina , Fator de Crescimento Insulin-Like I/farmacologia , Ilhotas Pancreáticas/citologia , Camundongos , Ratos , Especificidade da Espécie , Células-Tronco/citologia
9.
Hum Gene Ther ; 12(13): 1611-21, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11535165

RESUMO

Previously, we have demonstrated that chondroitin sulfate proteoglycans and glycosaminoglycans inhibit retrovirus transduction. While studying the mechanism of inhibition, we found that the combined addition of equal-weight concentrations (80 microg/ml) of Polybrene and chondroitin sulfate C to retrovirus stocks resulted in the formation of a high-molecular-weight retrovirus-polymer complex that could be pelleted by low-speed centrifugation. The pelleted complex contained more than 80% of the virus particles, but less than 0.3% of the proteins that were originally present in the virus stock. Surprisingly, the virus in the complex remained active and could be used to transduce cells. The titer of the pelleted virus, when resuspended in cell culture medium to the starting volume, was three-fold greater than the original virus stock. The selectivity (CFU/mg protein) of the process with respect to virus activity was more than 1000-fold. When the pelleted virus-polymer complex was resuspended in one-eighth of the original volume and used to transduce NIH 3T3 murine fibroblasts and primary human fibroblasts, gene transfer was increased 10- to 20-fold over the original unconcentrated retrovirus stock. The implications of our findings for the production, processing, and use of retrovirus stocks for human gene therapy protocols are discussed.


Assuntos
Terapia Genética/métodos , Polímeros/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Transdução Genética/métodos , Transgenes/genética , Células 3T3 , Animais , Ânions/metabolismo , Células CHO , Cátions/metabolismo , Centrifugação , Precipitação Química , Sulfatos de Condroitina/química , Sulfatos de Condroitina/metabolismo , Cricetinae , Fibroblastos/virologia , Genes Reporter/genética , Brometo de Hexadimetrina/química , Brometo de Hexadimetrina/metabolismo , Humanos , Camundongos , Peso Molecular , Especificidade de Órgãos , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Polímeros/química , Retroviridae/fisiologia
10.
Hum Gene Ther ; 9(4): 529-39, 1998 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9525314

RESUMO

Skin loss due to burns and ulcers is a major medical problem. Bioengineered skin substitutes that use cultured keratinocytes as an epidermal layer with or without analogues of the dermis are one strategy for skin repair. However, none can achieve definitive wound closure, function, or cosmesis comparable to split-thickness autografts. Moreover, autograft donor sites, which require time to heal, may be limited or have attendant problems such as infection or functional/cosmetic deficiencies. To determine if the performance of composite skin grafts of keratinocytes on a dermal analogue could be enhanced, human keratinocytes were genetically modified to overexpress platelet-derived growth factor A chain (PDGF-A). Composite grafts of modified keratinocytes seeded onto acellular dermis, prepared from cryopreserved cadaver skin, secreted PDGF-AA protein in vitro [90 ng/graft (1.5 x 1.5 cm)/24 hr]. To test their performance in a wound healing model, composite grafts were transplanted to full-thickness excisional wounds on the back of athymic mice. PDGF-A grafts formed a stratified differentiated epidermis similar to control grafts. The acellular dermis was repopulated with host fibrovascular cells and by day 7, the PDGF-A grafts had significantly more cells in the dermis and increased staining for murine collagen types I and IV. At this early time point, wound contraction was also significantly inhibited in PDGF-A grafts versus control grafts. Thus, PDGF-A overexpression improves graft performance during the first critical week after transplantation.


Assuntos
Sobrevivência de Enxerto , Queratinócitos/metabolismo , Queratinócitos/transplante , Fator de Crescimento Derivado de Plaquetas/genética , Transplante de Pele/métodos , Cicatrização , Células 3T3 , Animais , Células Cultivadas , Técnicas de Cocultura , Colágeno/análise , Técnicas de Cultura , Células Epidérmicas , Vetores Genéticos , Humanos , Queratinócitos/citologia , Óperon Lac , Camundongos , Camundongos Nus , Fator de Crescimento Derivado de Plaquetas/biossíntese , Retroviridae/genética , Pele/irrigação sanguínea , Pele/citologia
11.
Hum Gene Ther ; 8(5): 523-32, 1997 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-9095404

RESUMO

Because human fibroblasts are easily brought to tissue culture conditions and can be stably transduced with retroviral vectors encoding transgenes ex vivo, genetically modified fibroblasts are frequently considered in strategies to correct disease with gene therapy. This enthusiasm has been dampened by studies showing that transgene expression by genetically modified fibroblasts diminishes with time in vivo, but not in vitro, for reasons that are unclear. We elected to study this problem using cloned human fibroblasts that had been cloned by limiting dilution and stably transduced with a retroviral vector encoding lacZ ex vivo. These were seeded onto a nonbiodegradable nylon matrix that was transplanted to nude mice. Transgene expression was followed prospectively by histologic exam. Data show that human fibroblasts can withstand the pressure of cloning by limiting dilution. In addition, they can be passaged from 10 to > 20 times, and > 1 x 10(20) of genetically modified fibroblasts can be generated as progeny of one cell. Loss of transgene expression by the cloned genetically modified fibroblasts in vivo occurs in an orderly and progressive fashion, but is not complete by 4 months. Neither the loss nor the persistence of expression appear to be random. These observations are most compatible with the thesis that a major cause of the loss of transgene expression in vivo is secondary to apoptosis of the genetically modified fibroblast. Loss of expression of transgenes in senescent genetically modified fibroblasts occurs more rapidly than in their presenescent counterparts in the age-neutral, in vivo setting of the nude mouse.


Assuntos
Fibroblastos/fisiologia , Transdução Genética , Transgenes/genética , Animais , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Fibroblastos/citologia , Deleção de Genes , Expressão Gênica , Genes Reporter , Humanos , Óperon Lac/genética , Óperon Lac/fisiologia , Masculino , Camundongos , Camundongos Nus
12.
J Invest Dermatol ; 111(5): 810-6, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9804343

RESUMO

To aid in the investigation of factors that control the proliferation and function of melanocytes, we have characterized a skin equivalent model that supports melanocyte growth and function in vitro and in vivo. Passenger melanocytes survive and proliferate at low numbers when keratinocytes of the epidermis are cultured in serum-containing medium using a fibroblast feeder layer. When the surface of de-epidermalized acellular dermis was seeded with these cultured cells, the keratinocytes formed a stratified epithelium in vitro containing rete ridges, and the melanocytes were preferentially located in the bottom of these rete ridges. Melanocyte cell number was much less than in normal skin, but in some areas the melanocytes were in clusters, consistent with clonal growth of the cells. When transplanted to athymic mice, the grafts formed foci of pigmentation at 3 wk that expanded and repigmented the entire graft by 8 wk. Histologic examination of these foci revealed that they corresponded to clusters of melanocytes that proliferated and migrated to eventually repopulate the entire graft. In grafts of mixed cells from light and dark skin donors, distinct foci of pigmentation were obvious at 3 wk and, instead of progressing to complete repigmentation, these foci remained stable for over 6 wk. Histologic examination confirmed that these grafts of mixed cells were entirely repopulated with melanocytes and that the grafts contained distinct zones of melanocytes that were of exclusively dark or light skin origin. This model should be valuable for studying the clonal growth of melanocytes in the context of the epidermis.


Assuntos
Células Clonais/citologia , Melanócitos/citologia , Animais , Divisão Celular , Humanos , Recém-Nascido , Queratinócitos/citologia , Masculino , Camundongos , Modelos Biológicos , Pigmentação da Pele/fisiologia , Transplante de Pele/patologia
13.
J Invest Dermatol ; 112(3): 297-302, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10084305

RESUMO

Several techniques for cutaneous gene transfer have been investigated for either in vitro or in vivo applications. In the present study, we investigated whether the direct delivery of platelet-derived growth factor cDNA into skin results in improvement in tissue repair. Cutaneous transfections were carried out in rats using a particle-bombardment device (Accell). As revealed by reverse transcriptase-polymerase chain reaction, transgene expression in vivo was transient, with low level expression by day 5. When compared with wounds transfected with a control cytomegalovirus-luciferase plasmid, wounds transfected with platelet-derived growth factor A or B in the MFG vector showed a significant increase in wound tensile strength 7 and 14 d after transfection. At both time points platelet-derived growth factor A transfected wounds exhibited the highest increase in tensile strength over controls, resulting in a 3.5-fold increase at day 7 and a 1.5-fold increase at day 14. The degree of stimulation was not remarkably different between wounds transfected with platelet-derived growth factor B, which is predominantly cell associated, or a truncation mutant, platelet-derived growth factor B211, which is predominantly secreted. These findings demonstrate that in vivo gene transfer by particle bombardment can be used to improve the tissue repair response. This approach provides a robust tool to assess the biologic activity of various proteins and will aid in the development of therapeutic cutaneous gene delivery.


Assuntos
Técnicas de Transferência de Genes , Aceleradores de Partículas , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/fisiologia , Pele/lesões , Cicatrização/fisiologia , Animais , Expressão Gênica/fisiologia , Isomerismo , Masculino , Mutação/fisiologia , Ratos , Ratos Sprague-Dawley , Pele/fisiopatologia , Resistência à Tração , Transgenes/genética , Ferimentos e Lesões/fisiopatologia
14.
J Invest Dermatol ; 107(1): 113-20, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8752849

RESUMO

Somatomedin C/insulin-like growth factor-I (IGF-I) is required for the proliferation of keratinocytes in vitro. In skin, the cells known to synthesize IGF-I are melanocytes and fibroblasts of the dermis. To investigate the role of IGF-I as a mediator of keratinocyte proliferation, we have used retroviral-mediated gene transfer to introduce the gene encoding human IGF-I into diploid human keratinocytes, thus causing these cells to produce a growth factor they normally do not express. Modified cells synthesized and secreted significant levels of IGF-I (560 ng/10(7) cells/24 h) in vitro. Cells expressing IGF-I were no longer dependent on exogenously added IGF-I or insulin for their sustained growth in vitro under serum-free conditions. The growth of these cells did require added epidermal growth factor (EGF) and bovine pituitary extract. The addition of an antibody that neutralizes IGF-I inhibited cell growth, suggesting that IGF-I must be secreted by the cells to promote cell proliferation. To investigate the role of IGF-I in vivo, we grafted modified keratinocytes expressing IGF-I onto athymic mice. Grafts of epithelial sheets of modified cells formed a stratified epithelium comparable to control grafts of unmodified cells. When analyzed for keratin 16 expression and by quantitative staining for the nuclear proliferation antigen Ki-67, however, modified epithelia showed an increase in these markers of proliferation when compared with grafts of unmodified cells. This study demonstrates that genetic modification can be used to modify the autocrine control of keratinocyte proliferation. The de novo synthesis of IGF-I by keratinocytes could sustain keratinocytes growth in vitro and stimulate proliferation in vivo without significantly altering epidermal differentiation. These data further support the role of IGF-I as a paracrine mediator of epidermal proliferation and as a potential signal of mesenchymal-epithelial interactions.


Assuntos
Marcação de Genes , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Animais , Sequência de Bases , Divisão Celular , Células Cultivadas , Células Epiteliais , Técnicas de Transferência de Genes , Humanos , Queratinócitos/transplante , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Sondas de Oligonucleotídeos/genética
15.
J Invest Dermatol ; 114(4): 647-53, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10733668

RESUMO

We examined the role of the serine proteinase plasmin in regulating fibroblast-mediated tissue remodeling during wound healing. As an in vitro model system, collagen lattices were seeded with human dermal fibroblasts, and various concentrations of plasmin were added to the medium of the contracting lattices. Within 16 h, fibroblast-populated collagen lattices treated with plasmin rapidly contracted from approximately 20 mm to less than 2 mm in diameter. Measurements of collagen lattices with radiolabeled collagen indicated that, when these lattices included either fibroblasts or conditioned medium derived from fibroblast-populated collagen lattices, exogenous plasmin induced collagen degradation and rapid lattice contraction. Western blot analyses of conditioned medium demonstrated that fibroblasts in collagen lattices secreted the latent matrix metalloproteinase, MMP-1, which was subsequently cleaved by plasmin. Additionally, rapidlattice contraction and collagen degradation were blocked when collagen lattices were treated simultaneously with plasmin and aprotinin or a tissue inhibitor of metalloproteinases, TIMP-1. These results provide strong evidence that plasmin regulates rapid contraction of collagen lattices by activating fibroblast-secreted MMP-1 that triggers collagen degradation. The findings from this study suggest that fibroblast-populated collagen lattices can be used as an in vitro model system to investigate the mechanisms by which plasmin and cell-secreted plasminogen activators control MMP-1 mediated extracellular lattice degradation and remodeling during wound healing.


Assuntos
Colágeno/metabolismo , Fibrinolisina/fisiologia , Fibroblastos/fisiologia , Aprotinina/farmacologia , Linhagem Celular , Fibroblastos/metabolismo , Humanos , Recém-Nascido , Masculino , Metaloproteinase 1 da Matriz/metabolismo , Modelos Biológicos , Inibidores de Proteases/farmacologia , Inibidores de Serina Proteinase/farmacologia , Inibidor Tecidual de Metaloproteinase-1/farmacologia , Cicatrização
16.
J Invest Dermatol ; 112(2): 233-9, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9989801

RESUMO

Cells transduced ex vivo with transgenes encoded on retroviruses have constant and prolonged expression in vitro; however, in vivo expression is quickly lost. Much attention has been directed at methods to circumvent this problem. We have shown that loss of transgene expression does not occur when transduced immortalized 3T3 cells are transplanted to the in vivo setting of athymic mice. Ease of acquisition and potential for clinical application led us to assess the potential of using immortalized human keratinocytes for expression of transgenes in vivo. Human keratinocytes were immortalized with a HPV16-E6/E7 retrovirus, transduced with a lacZ retrovirus, cloned by limiting dilution, seeded onto a physiologic dermal substrate, and transplanted to athymic mice. Six weeks after transplantation, the immortalized transgene expressing keratinocytes had formed an epidermis that was indistinguishable from one formed by nonimmortalized keratinocytes; furthermore, there was no loss of expression of the lacZ gene. These observations show that methods to extend cell survival are an alternative approach to achieving stable and prolonged expression of transgenes in vivo and that HPV16-E6/ E7 immortalized keratinocytes generate an epidermis with normal morphology.


Assuntos
Queratinócitos/citologia , Transgenes/genética , Animais , Diferenciação Celular/genética , Sobrevivência Celular/genética , Cromossomos Humanos Par 10/genética , Cromossomos Humanos Par 9/genética , Expressão Gênica , Humanos , Cariotipagem , Óperon Lac/genética , Masculino , Camundongos , Camundongos Nus , Proteínas Oncogênicas Virais , Papillomaviridae , Transdução Genética
17.
J Invest Dermatol ; 104(2): 171-6, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7829871

RESUMO

Somatic gene therapy has been proposed as a means of treating inherited diseases involving defective or absent plasma proteins, viral diseases, and cancer. Introduction of the gene of interest into fibroblasts and implantation of these genetically modified fibroblasts using a skin equivalent system may be an attractive model for gene therapy because skin fibroblasts are easily obtained and propagated in culture. This study evaluated expression of the gene for human transferrin (hTf) by genetically modified fibroblasts in vitro and in vivo. NIH 3T3 fibroblasts, which form non-metastasizing tumors in athymic mice, were transduced with a retroviral vector encoding hTf. The transduced cells were cloned by limiting dilution and hTf production by the cloned cells measured. Two clones of cells producing high levels of hTf were used to seed collagen-coated nylon matrices, which were maintained in culture for up to 53 d. The rate of synthesis of hTf by the seeded matrices was constant after 22 d in vitro. Matrices seeded with cloned, transduced cells were implanted subcutaneously into seven athymic mice, and plasma levels of hTf were assessed biweekly. In all animals, the plasma level of hTf was detectable at week 6 after implantation. Levels of hTf remained elevated in the animals until the implants were removed at week 12. At week 10, the level of hTf in the plasma correlated with tumor volume in tumors less than 2000 mm3 in size. The half-life of hTf in the mice was 39.5 h. In this model, gene expression did not decline for the 12-week observation period.


Assuntos
Terapia Genética , Transferrina/genética , Transferrina/metabolismo , Células 3T3/transplante , Animais , Células Clonais/química , Células Clonais/metabolismo , Colágeno/análise , Humanos , Camundongos , Camundongos Nus , Recombinação Genética , Retroviridae/genética , Transplante de Pele/fisiologia , Transdução Genética
18.
J Invest Dermatol ; 107(1): 121-7, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8752850

RESUMO

This study evaluates the use of composite grafts of cultured human keratinocytes and de-epidermalized, acellular human dermis to close full-thickness wounds in athymic mice. Grafts were transplanted onto athymic mice and studied up to 8 wk. Graft take was excellent, with no instances of infection or graft loss. By 1 wk, the human keratinocytes had formed a stratified epidermis that was fused with mouse epithelium, and by 8 wk the grafts resembled human skin and could be freely moved over the mouse dorsum. Immunostaining for keratins 10 and 16 and for involucrin revealed an initial pattern of epithelial immaturity, which by 8 wk had normalized to that of mature unwounded epithelium. Mouse fibroblasts began to infiltrate the acellular dermis as early as 1 wk. By 8 wk fibroblasts had completely repopulated the dermis, and blood vessels were evident in the most superficial papillary projections. Dermal elements, such as rete ridges and elastin fibers, which were present in the starting dermis, persisted for the duration of the experiment. Grafts using keratinocytes from dark-skinned donors as opposed to light-skin donors had foci of pigmentation as early as 1 wk that progressed to homogenous pigmentation of the graft by 6 wk. These results indicate that melanocytes that persist in vitro are able to resume normal function in vivo. Our study demonstrates that composite grafts of cultured keratinocytes combined with acellular dermis are a useful approach for the closure of full-thickness wounds.


Assuntos
Queratinócitos/transplante , Fenômenos Fisiológicos da Pele , Transplante de Pele , Animais , Células Cultivadas , Células Epidérmicas , Fibroblastos/citologia , Humanos , Imuno-Histoquímica , Queratinócitos/citologia , Queratinócitos/fisiologia , Camundongos , Camundongos Nus , Neovascularização Fisiológica , Pele/irrigação sanguínea
19.
J Invest Dermatol ; 105(6): 756-63, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7490468

RESUMO

We investigated the hypothesis that keratinocyte-produced platelet-derived growth factor-AA (PDGF-AA) is involved in epidermal-dermal interactions and that PDGF-AA is an important mediator of the temporal and spatial events of tissue repair. Retroviral-mediated gene transfer was used to introduce the gene encoding human PDGF-A into cultures of human diploid keratinocytes. Genetic modification boosted the endogenous in vitro level of PDGF-AA secretion by over 300 fold. When PDGF-secreting cells were transplanted as epithelial sheets to athymic mice, modified keratinocytes underwent terminal differentiation and generated a stratified epithelium comparable to unmodified cells. Seven days after grafting the newly synthesized connective tissue layer subjacent to the PDGF-A-modified grafts was significantly thicker, was rich in mononuclear cells and fibroblasts, and had increased numbers of blood vessels when compared to control grafts of unmodified cells. These results suggest that PDGF-AA secreted by the epidermis is an important mediator of epithelial-mesenchymal interactions and helps to promote growth and vascularization of the underlying dermal tissue. Further, these data demonstrate the feasibility of using genetically modified cells to modulate tissue regeneration.


Assuntos
Tecido Conjuntivo/fisiologia , Queratinócitos/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Regeneração , Animais , Células Cultivadas , Tecido Conjuntivo/irrigação sanguínea , Humanos , Queratinócitos/transplante , Camundongos , Camundongos Nus , Neovascularização Fisiológica , Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Recombinantes/biossíntese , Células Estromais/fisiologia
20.
J Invest Dermatol ; 103(5 Suppl): 76S-84S, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7963689

RESUMO

Molecular definition of disease at the level of the gene and advances in recombinant DNA technology suggest that many diseases are amenable to correction by genes not bearing the defective elements that result in disease. Many questions must be answered before this therapy can be used to correct chronic diseases. These questions fall into safety and efficacy categories. Experience with transplanting cellular elements of skin or skin substitutes (defined as skin that possess the cell types and a dermal structure to develop into a functioning skin) to athymic rodents is considerable and is seen as a system where these questions can be answered. This paper reviews these questions and presents our early analysis of genetically modified cells in skin substitutes in vivo and in vitro. Experimental data demonstrate that both a matrix of woven nylon, housing a fibroblast generated collage, and dead dermis can be utilized to shuttle genetically modified human fibroblasts from the laboratory to an in vivo setting. Genetically modified fibroblasts do not migrate from the shuttle to the surrounding tissue. The survival of significant numbers, approximately 70%, of genetically modified fibroblasts for at least 6 weeks in these shuttles, supports this general approach as having clinical utility. It is also concluded that skin substitute systems can be used to generate a genetically modified skin in vitro that has the capacity to develop into functional skin in vivo. Further, as genetically modified keratinocytes differentiate there is increased production by the transgene, supporting the concept that keratinocytes have true potential as shuttles for therapeutic genes. This work demonstrates that transplantation of systems containing genetically modified cells of the skin can be used to experimentally define many aspects of gene therapy using skin before this technology is taken to the clinic. Examples include determining the effect of gene transduction and expression on structure and function of the genetically modified skin as well as on distant skin and an assessment of the translational capacity of the transgene as function of time and cell number.


Assuntos
Terapia Genética/métodos , Pele , Células 3T3 , Abdome , Animais , Gonadotropina Coriônica/metabolismo , Estudos de Avaliação como Assunto , Fibroblastos , Técnicas Histológicas , Humanos , Recém-Nascido , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pênis , Pele/citologia , Transplante de Pele
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA