Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
2.
Eur Heart J ; 39(Suppl 1)2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30270958

RESUMO

Introduction: Focussing on the potential role of cardiovascular cell therapy, we investigated the spatial relationship between pericytes (cells with cardiac repair capabilities that ensheath blood vessels) and endogenous cardiac progenitors within stem cells' niches. We explored possible changes in their co-localisation in developing human hearts from foetal to adult stage and following ischaemia. Methods: Foetal and adult human heart specimens, obtained under ethical consent (University of Edinburgh ethics committee), were used for immunohistochemistry, cell isolation, culture and differentiation. Multi-lineage differentiation in culture, by single and double staining was completed for CD 146+ foetal pericytes and c-kit+ cells. Endothelial markers (CD31) gene expression was quantified by qPCR. Results: c-kit+ cells frequency and coexpression with pericytes decrease with heart development, already evident by gestation week 19th. Pericytes and c-kit+ cells express the early cardiac transcription factors Nkx2.5 and Islet 1. Only c-kit+ cells express the stemness marker SSEA3 (24%), known to progressively decrease with cell differentiation. Endothelial differentiation assessment shows that cardiac pericytes and c-kit+ cells do not form CD31+ networks. This finding correlates with absence of staining for CD31 marker in both cultured cells' types. The cardiac marker α-actin was present in both cell populations. In healthy adult heart, pericyte markers CD146 localise within the vasculature. Following ischaemia this pericyte marker becomes also evident outside the vasculature.In healthy adult atrium, c-kit expression is low and coexpression with other markers inconspicuous. Ischaemia leads to increased c-kit expression, particularly in blood vessels <50um diameter. Furthermore, following ischaemia c-kit, endothelium and pericyte markers co-localise within the same atrial cells. Blood vessels >50µm diameter showed mostly only staining for endothelial (vWF) and pericyte (CD146) markers, with no co-expression of c-kit marker identified. Staining patterns within the ischaemic regions of the right and left atrial appendages revealed low levels of colocalisation between vWF and CD146. Acute ischaemia of the left ventricle affected the detection of cardiac stem cells markers in the area of injury, due to myocardium disruption. Conclusion: Foetal heart pericytes and c-kit+ cells express early cardiac transcription factors and show trans-differentiation potential, which decreases in healthy adult hearts. The preservation and activity of cardiac stem cells niches within the atrium vasculature appears re-activated in post-ischaemic hearts. Better understanding of cardiac c-kit+ and pericyte cells during-human embryonic development and during ischaemia may identify alternative novel therapeutic strategy against coronary artery disease.

3.
Nat Commun ; 8(1): 1118, 2017 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-29061963

RESUMO

Mesenchymal cells expressing platelet-derived growth factor receptor beta (PDGFRß) are known to be important in fibrosis of organs such as the liver and kidney. Here we show that PDGFRß+ cells contribute to skeletal muscle and cardiac fibrosis via a mechanism that depends on αv integrins. Mice in which αv integrin is depleted in PDGFRß+ cells are protected from cardiotoxin and laceration-induced skeletal muscle fibrosis and angiotensin II-induced cardiac fibrosis. In addition, a small-molecule inhibitor of αv integrins attenuates fibrosis, even when pre-established, in both skeletal and cardiac muscle, and improves skeletal muscle function. αv integrin blockade also reduces TGFß activation in primary human skeletal muscle and cardiac PDGFRß+ cells, suggesting that αv integrin inhibitors may be effective for the treatment and prevention of a broad range of muscle fibroses.


Assuntos
Integrina alfaV/metabolismo , Músculo Esquelético/patologia , Miocárdio/patologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Animais , Apoptose , Movimento Celular , Células Cultivadas , Colágeno/metabolismo , Fibrose , Genótipo , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes/metabolismo
4.
Stem Cell Res Ther ; 7: 47, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-27029948

RESUMO

BACKGROUND: Adipose tissue is an attractive source of mesenchymal stem cells (MSC) as it is largely dispensable and readily accessible through minimally invasive procedures such as liposuction. Until recently MSC could only be isolated in a process involving ex-vivo culture and their in-vivo identity, location and frequency remained elusive. We have documented that pericytes (CD45-, CD146+, and CD34-) and adventitial cells (CD45-, CD146-, CD34+) (collectively termed perivascular stem cells or PSC) represent native ancestors of the MSC, and can be prospectively purified using fluorescence activated cell sorting (FACS). In this study we describe an optimized protocol that aims to deliver pure, viable and consistent yields of PSC from adipose tissue. We analysed the frequency of PSC within adipose tissue, and the effect of patient and procedure based variables on this yield. METHODS: Within this twin centre study we analysed the adipose tissue of n = 131 donors using flow cytometry to determine the frequency of PSC and correlate this with demographic and processing data such as age, sex, BMI and cold storage time of the tissue. RESULTS: The mean number of stromal vascular fraction (SVF) cells from 100 ml of lipoaspirate was 34.4 million. Within the SVF, mean cell viability was 83 %, with 31.6 % of cells being haematopoietic (CD45+). Adventitial cells and pericytes represented 33.0 % and 8 % of SVF cells respectively. Therefore, a 200 ml lipoaspirate would theoretically yield 23.2 million viable prospectively purified PSC - sufficient for many reconstructive and regenerative applications. Minimal changes were observed in respect to age, sex and BMI suggesting universal potential application. CONCLUSIONS: Adipose tissue contains two anatomically and phenotypically discreet populations of MSC precursors - adventitial cells and pericytes - together referred to as perivascular stem cells (PSC). More than 9 million PSC per 100 ml of lipoaspirate can be rapidly purified to homogeneity using flow cytometry in clinically relevant numbers potentially circumventing the need for purification and expansion by culture prior to clinical use. The number and viability of PSC are minimally affected by patient age, sex, BMI or the storage time of the tissue, but the quality and consistency of yield can be significantly influenced by procedure based variables.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Adulto , Antígenos CD/metabolismo , Separação Celular , Células Cultivadas , Demografia , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Pericitos/metabolismo , Estudos Prospectivos , Gordura Subcutânea/citologia , Preservação de Tecido , Adulto Jovem
5.
Sci Rep ; 6: 22779, 2016 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-26997456

RESUMO

Atrophic non-union is attributed to biological failure of the fracture repair process. It occurs in up to 10% of fractures, results in significant morbidity to patients, and treatment often requires complex reconstructive procedures. We tested the ability of human bone derived marrow mesenchymal stem cells (MSC), and human adipose derived pericytes (the native ancestor of the MSC) delivered percutaneously to the fracture gap to prevent the formation of atrophic non-union in a rat model. At eight weeks, 80% of animals in the cell treatment groups showed evidence of bone healing compared to only 14% of those in the control group. Radiographic parameters showed significant improvement over the eight-week period in the cell treatment groups, and histology confirmed bone bridges at the fracture gap in the both treatment groups. The quality of bone produced and its biomechanical properties were significantly enhanced in both treatment groups. The results from this study demonstrate that MSC and pericytes have significant bone regeneration potential in an atrophic non-union model. These cells may have a role in the prevention of atrophic non-union and could enable a paradigm shift in the treatment of fractures at high risk of failing to heal and developing non-union.


Assuntos
Transplante de Células-Tronco Mesenquimais , Pericitos/transplante , Fraturas da Tíbia/terapia , Tecido Adiposo Branco/citologia , Animais , Feminino , Consolidação da Fratura , Humanos , Células-Tronco Mesenquimais/fisiologia , Pericitos/fisiologia , Ratos Wistar , Tíbia/diagnóstico por imagem , Tíbia/fisiopatologia , Fraturas da Tíbia/diagnóstico por imagem
6.
Stem Cell Rev Rep ; 10(6): 830-40, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24943487

RESUMO

Duchenne muscular dystrophy (DMD) is still an untreatable lethal X-linked disorder, which affects 1 in 3500 male births. It is caused by the absence of muscle dystrophin due to mutations in the dystrophin gene. The potential regenerative capacity as well as immune privileged properties of mesenchymal Stem Cells (MSC) has been under investigation for many years in an attempt to treat DMD. One of the questions to be addressed is whether stem cells from distinct sources have comparable clinical effects when injected in murine or canine muscular dystrophy animal models. Many studies comparing different stem cells from various sources were reported but these cells were obtained from different donors and thus with different genetic backgrounds. Here we investigated whether human pericytes obtained from 4 different tissues (muscle, adipose tissue, fallopian tube and endometrium) from the same donor have a similar clinical impact when injected in double mutant Utrn (tm1Ked) Dmd (mdx) /J mice, a clinically relevant model for DMD. After a weekly regimen of intraperitoneal injections of 10(6) cells per 8 weeks we evaluated the motor ability as well as the life span of the treated mice as compared to controls. Our experiment showed that only adipose tissue derived pericytes are able to increase significantly (39 days on average) the life span of affected mice. Microarray analysis showed an inhibition of the interferon pathway by adipose derived pericytes. Our results suggest that the clinical benefit associated with intraperitoneal injections of these adult stem cells is related to immune modulation rather than tissue regeneration.


Assuntos
Tecido Adiposo/fisiologia , Pericitos/fisiologia , Tecido Adiposo/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Distrofina/metabolismo , Feminino , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos mdx , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Pericitos/metabolismo
7.
Bone Joint J ; 96-B(3): 291-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24589781

RESUMO

The ability of mesenchymal stem cells (MSCs) to differentiate in vitro into chondrocytes, osteocytes and myocytes holds great promise for tissue engineering. Skeletal defects are emerging as key targets for treatment using MSCs due to the high responsiveness of bone to interventions in animal models. Interest in MSCs has further expanded in recognition of their ability to release growth factors and to adjust immune responses. Despite their increasing application in clinical trials, the origin and role of MSCs in the development, repair and regeneration of organs have remained unclear. Until recently, MSCs could only be isolated in a process that requires culture in a laboratory; these cells were being used for tissue engineering without understanding their native location and function. MSCs isolated in this indirect way have been used in clinical trials and remain the reference standard cellular substrate for musculoskeletal engineering. The therapeutic use of autologous MSCs is currently limited by the need for ex vivo expansion and by heterogeneity within MSC preparations. The recent discovery that the walls of blood vessels harbour native precursors of MSCs has led to their prospective identification and isolation. MSCs may therefore now be purified from dispensable tissues such as lipo-aspirate and returned for clinical use in sufficient quantity, negating the requirement for ex vivo expansion and a second surgical procedure. In this annotation we provide an update on the recent developments in the understanding of the identity of MSCs within tissues and outline how this may affect their use in orthopaedic surgery in the future.


Assuntos
Células do Tecido Conjuntivo/fisiologia , Células-Tronco Mesenquimais/fisiologia , Ortopedia , Engenharia Tecidual/métodos , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Humanos
8.
J Plast Reconstr Aesthet Surg ; 67(6): 745-51, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24529696

RESUMO

Access to human tissue is critical to medical research, however the laws and regulations surrounding gaining ethical and legal access to tissue are often poorly understood. Recently, there has been a huge increase in the interest surrounding the therapeutic application of adipose tissue, and adipose-derived stem cells. To facilitate our own research interests and possibly assist our local colleagues and collaborators, we established a Research Tissue Bank (RTB) to collect, store and distribute human adipose tissue derived cells with all the appropriate ethical approval for subsequent downstream research. Here we examine the legal, ethical and practical issues relating to the banking of adipose tissue for research in the UK, and discuss relevant international guidelines and policies. We also share our experiences of establishing an RTB including the necessary infrastructure and the submission of an application to a Research Ethics Committee (REC).


Assuntos
Pesquisa Biomédica , Transplante de Células-Tronco/ética , Transplante de Células-Tronco/legislação & jurisprudência , Bancos de Tecidos/ética , Bancos de Tecidos/legislação & jurisprudência , Adipócitos/transplante , Comitês de Ética em Pesquisa/organização & administração , Feminino , Humanos , Masculino , Células-Tronco , Preservação de Tecido , Obtenção de Tecidos e Órgãos/ética , Obtenção de Tecidos e Órgãos/legislação & jurisprudência
9.
Pflugers Arch ; 465(6): 767-73, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23588377

RESUMO

Pericytes have become a hot topic in renal biology. They play a critical physiological role in vessel development, maintenance and remodelling through active communication with their vascular partners-endothelial cells-and modulation of extracellular matrix proteins. Multiple functions for renal pericytes have been described; specialised perivascular populations participate in glomerular filtration, regulate medullary blood flow and contribute to kidney fibrosis by differentiation into collagen-generating myofibroblasts. Interestingly, the origin of renin-producing cells of the juxtaglomerular region is attributed to the perivascular cell lineage; we have observed the coincidence of renin and pericyte marker expression during human kidney development. Finally, pericytes have been shown to share features with mesenchymal stem cells, which places them as potential renal progenitor cell candidates. Since renal diseases are often associated with microvascular complications, renal pericytes may emerge as new targets for the treatment of kidney disease.


Assuntos
Rim/citologia , Pericitos/fisiologia , Animais , Humanos , Pericitos/citologia , Pericitos/metabolismo
10.
Int J Obes (Lond) ; 36(1): 155-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21522126

RESUMO

Brown adipose tissue mitochondria express the unique thermogenic uncoupling protein-1. Recently, brown adipocyte progenitors have been identified in the CD34+ cell population of human skeletal muscle. The aims of this study were firstly to determine if obesity and diabetes have altered amounts of muscle brown adipocyte progenitors and, secondly, to establish if the latter are correlated with clinical parameters of obesity and diabetes. Body mass index (BMI), plasma glucose, insulin, cholesterol and triglycerides as well as homeostasis model assessment were measured in lean (n=10), obese (n=18) and obese-diabetic (n=15) subjects and muscle biopsies were taken from the rectus abdominus. CD34 being also expressed on endothelial cells, we measured CD31, another endothelial marker, and expressed the brown adipocyte progenitors, as the CD34/CD31 mRNA ratio. The latter was significantly reduced in the obese vs lean subjects suggesting a smaller pool of brown adipocyte progenitors. More strikingly, for lean and obese subjects negative correlations were observed between the CD34/CD31 mRNA ratios and BMI, fasting insulin levels and homeostasis model assessment. These correlations highlight the potential physiological relevance of the muscle CD34/CD31 mRNA ratio.


Assuntos
Adipócitos Marrons/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Músculo Esquelético/metabolismo , Obesidade/metabolismo , Células-Tronco/metabolismo , Magreza/metabolismo , Adulto , Antígenos CD34/genética , Antígenos CD34/metabolismo , Glicemia/metabolismo , Índice de Massa Corporal , Colesterol/sangue , Diabetes Mellitus Tipo 2/diagnóstico por imagem , Feminino , Humanos , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/diagnóstico por imagem , Obesidade/diagnóstico por imagem , Obesidade/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Reação em Cadeia da Polimerase/métodos , RNA Mensageiro/metabolismo , Cintilografia , Triglicerídeos/sangue
12.
J Exp Med ; 197(6): 763-75, 2003 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-12642604

RESUMO

A functional hybrid receptor associating the common gamma chain (gammac) with the granulocyte/macrophage colony-stimulating factor receptor beta (GM-CSFRbeta) chain is found in mobilized human peripheral blood (MPB) CD34+ hematopoietic progenitors, SCF/Flt3-L primed cord blood (CB) precursors (CBPr CD34+/CD56-), and CD34+ myeloid cell lines, but not in normal natural killer (NK) cells, the cytolytic NK-L cell line or nonhematopoietic cells. We demonstrated, using CD34+ TF1beta cells, which express an interleukin (IL)-15Ralpha/beta/gammac receptor, that within the hybrid receptor, the GM-CSFRbeta chain inhibits the IL-15-triggered gammac/JAK3-specific signaling controlling TF1beta cell proliferation. However, the gammac chain is part of a functional GM-CSFR, activating GM-CSF-dependent STAT5 nuclear translocation and the proliferation of TF1beta cells. The hybrid receptor is functional in normal hematopoietic progenitors in which both subunits control STAT5 activation. Finally, the parental TF1 cell line, which lacks the IL-15Rbeta chain, nevertheless expresses both a functional hybrid receptor that controls JAK3 phosphorylation and a novel IL-15alpha/gammac/TRAF2 complex that triggers nuclear factor kappaB activation. The lineage-dependent distribution and function of these receptors suggest that they are involved in hematopoiesis because they modify transduction pathways that play a major role in the differentiation of hematopoietic progenitors.


Assuntos
Antígenos CD34/metabolismo , Células-Tronco Hematopoéticas/imunologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Receptores de Interleucina-2/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos Monoclonais/metabolismo , Divisão Celular/fisiologia , Linhagem Celular , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Células Matadoras Naturais/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Receptores de Interleucina-15 , Receptores de Interleucina-2/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais/fisiologia
13.
Diabetologia ; 44(11): 2066-76, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11719839

RESUMO

AIMS/HYPOTHESIS: Transplanting human pancreatic islet beta cells could represent a radical new treatment of Type I (insulin-dependent) diabetes mellitus. However, beta cells available for grafting are scarce and finding new sources of such cells would be crucial for any cell therapy for diabetes. Undifferentiated precursor cells present in the human embryonic pancreas could represent such a source. METHODS: We grafted human embryonic pancreases (6-9 weeks of development) that contain very few beta cells onto NOD/scid mice. RESULTS: The human pancreatic tissue grew, increasing in weight 200 times within six months and endocrine cells differentiated, the number of human beta cells being increased by a factor 5000. Finally, the developed human endocrine tissue was mature enough to control the glycaemia of mice deficient in endogenous beta cells. CONCLUSION/INTERPRETATION: Human embryonic pancreas represent a source of immature cells that can proliferate and differentiate into mass beta cells after transplantation. Transplantation of human embryonic pancreas into NOD/scid mice is a useful model for understanding the development of the human pancreas during prenatal life.


Assuntos
Glicemia/metabolismo , Transplante de Tecido Fetal/fisiologia , Transplante de Pâncreas/fisiologia , Animais , Idade Gestacional , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos SCID , Tamanho do Órgão , Pâncreas/crescimento & desenvolvimento , Valores de Referência
14.
Immunity ; 15(3): 487-95, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11567638

RESUMO

We have traced emerging hematopoietic cells along human early ontogeny by culturing embryonic tissue rudiments in the presence of stromal cells that promote myeloid and B cell differentiation, and by assaying T cell potential in the NOD-SCID mouse thymus. Hematogenous potential was present inside the embryo as early as day 19 of development in the absence of detectable CD34+ hematopoietic cells, and spanned both lymphoid and myeloid lineages from day 24 in the splanchnopleural mesoderm and derived aorta where CD34+ progenitors appear at day 27. By contrast, hematopoietic cells arising in the third week yolk sac, as well as their progeny at later stages, were restricted to myelopoiesis and therefore are unlikely to contribute to definitive hematopoiesis in man.


Assuntos
Embrião de Mamíferos/citologia , Células-Tronco Hematopoéticas/fisiologia , Mesoderma/citologia , Saco Vitelino/citologia , Antígenos CD34/análise , Aorta/citologia , Linhagem da Célula , Feminino , Hematopoese , Humanos , Gravidez
15.
Blood ; 96(12): 3763-71, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11090058

RESUMO

Development of the full repertoire of hematopoietic-lymphopoietic cells from a single stem cell requires specific contacts with stromal cells. The spatio-temporal organization of these cell associations in the bone marrow in ontogeny is, however, not well understood. In the adult, 10% of marrow cells form a cohort of compact aggregates, the hematon. In the hematon mesenchymal cells (Stro-1(+)), perivascular lipocytes (desmin(+)), endothelial cells (CD34(+), Flk-1(+), Sca-1(+)), and macrophages amalgamate with the hematopoietic progenitors long-term culture-initiating cells (LTC-IC), cobblestone area-forming cell (CAFC), high-proliferative-potential colony-forming unit (HPP-CFU), granulocyte-macrophage (GM)-CFU, and burst-forming unit-erythroid (BFU-E). During endochondral ossification of the femur, GM-CFU and day 7 CAFC numbers increased progressively from day 17 of gestation, but primitive, day 35 LTC-IC appeared from postnatal day 2. Unexpectedly, bone marrow (BM) taken between embryonic day 17 and day 5 was unable to support myeloid cell production in long-term cultures or to support day 35 LTC-IC growth. However, a gain in stromal cell competence occurred between days 7 and 10, which was correlated with the emergence of hematon in the BM. Thus, acquisition of hematopoietic competence by BM lags behind for approximately 10 days after the initial hematopoietic cell influx. In the adult, the hematon fraction was 3.7-fold enriched in day 35 LTC-IC over the buffy coat. It produced more GM-CFU and HPP-CFU in myeloid culture and more B cells in lymphopoietic "switch" cultures. It is reported that stromal hematopoietic units named hematons are specific morphogenetic structures that emerge at a well-defined postnatal stage of development in long bones, delineate discrete territories for hematopoietic stem cell seeding and development, embody the most productive hematogenous compartment in the BM, and probably enclose a morphogenetic organizer. (Blood. 2000;96:3763-3771)


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Hematopoéticas/citologia , Células Estromais/citologia , Animais , Linfócitos B/citologia , Desenvolvimento Ósseo/fisiologia , Medula Óssea/embriologia , Medula Óssea/crescimento & desenvolvimento , Medula Óssea/fisiologia , Adesão Celular/fisiologia , Agregação Celular/fisiologia , Comunicação Celular/fisiologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Embrião de Mamíferos , Fêmur/anatomia & histologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Imuno-Histoquímica , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese , Células Estromais/fisiologia
17.
Am J Respir Cell Mol Biol ; 23(2): 121-7, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10919974

RESUMO

Exacerbated inflammation is now recognized as an important component of cystic fibrosis (CF) airway disease. Whether inflammation is part of the basic defect in CF or a response to persistent infection remains controversial. We addressed this question using human fetal tracheal grafts in severe combined immunodeficient mice. This model yields histologically mature, and most importantly, naive CF and non-CF surrogate airways. Significant inflammatory imbalance was found in naive CF airway grafts, including a highly increased intraluminal interleukin 8 content (CF: 10.1 +/- 2.2 ng/ml; non-CF: 1.2 +/- 0.6 ng/ml; P < 0.05) and consistent accumulation of leukocytes in the subepithelial region (P < 0.001). CF airway grafts were not histologically affected until challenged with Pseudomonas aeruginosa, which provoked: (1) early (before 3 h) and massive leukocyte transepithelial migration, (2) intense epithelial exfoliation, and (3) rapid progression of bacteria toward the lamina propria. In non-CF grafts, these three sets of events were not observed before 6 h. Using a model of naive human airways, we thus demonstrate that before any infection, CF airways are in a proinflammatory state. After infection, the basal inflammatory imbalance contributes to exert severe damage to the mucosa, paving the way for bacterial colonization and subsequent steps of CF airway disease.


Assuntos
Fibrose Cística/patologia , Transplante de Tecido Fetal , Traqueia/transplante , Transplante Heterólogo/patologia , Animais , Líquido da Lavagem Broncoalveolar/química , Fibrose Cística/embriologia , Fibrose Cística/metabolismo , Feto , Humanos , Inflamação/patologia , Interleucina-8/metabolismo , Leucócitos/patologia , Camundongos , Camundongos SCID , Microscopia Eletrônica , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Traqueia/embriologia , Traqueia/ultraestrutura
18.
J Hematother Stem Cell Res ; 9(2): 175-81, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10813530

RESUMO

Transplantation of BM stromal cells engineered to secrete therapeutic factors could represent a treatment for a large array of hematologic disorders. The aim of this study was to evaluate the susceptibility of human BM stromal cell precursors to retroviral gene transfer, then the ability of those to be transplanted in vivo. We have transduced a recombinant retrovirus encoding the mouse CD2 antigen into STRO-1+ cells selected from adult and fetal BM. Gene-modified stromal cells were injected intravenously into NOD-SCID mice engrafted previously with pieces of human fetal hematopoietic bone. Using nested PCR, transgenic human cells were detected both in the marrow of human bone grafts and in the BM, liver, and spleen of host mice 7 weeks after grafting. These data indicate that BM stromal progenitor cells are targets for retrovirus-mediated gene transfer and can home to hematopoietic tissues on engraftment through the bloodstream of nonconditioned hosts.


Assuntos
Transplante de Medula Óssea , Terapia Genética/métodos , Transplante Heterólogo , Adulto , Animais , Células da Medula Óssea/imunologia , Transplante Ósseo/métodos , Técnicas de Cultura de Células , Transplante de Tecido Fetal/métodos , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Estromais/imunologia , Células Estromais/transplante , Distribuição Tecidual , Transfecção , Transgenes , Quimeras de Transplante
19.
Blood ; 95(10): 3113-24, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10807777

RESUMO

Three distinct classes of epitopes on human CD164 have been identified. Two of these, recognized by the monoclonal antibodies 105A5 and 103B2/9E10, are the CD164 class I and class II functionally defined epitopes, which cooperate to regulate adhesion and proliferation of CD34(+) cell subsets. In this article, we demonstrate that these 2 CD164 epitopes are expressed on CD34(+) cells throughout ontogeny, in particular on CD34(+ )cell clusters associated with the ventral floor of the dorsal aorta in the developing embryo and on CD34(+) hematopoietic precursor cells in fetal liver, cord blood, and adult bone marrow. While higher levels of expression of these CD164 epitopes occur on the more primitive AC133(hi)CD34(hi)CD38(lo/-) cell population, they also occur on most cord blood Lin(-)CD34(lo/-)CD38(lo/- )cells, which are potential precursors for the AC133(hi)CD34(hi)CD38(lo/-) subset. In direct contrast to these common patterns of expression on hematopoietic precursor cells, notable differences in expression of the CD164 epitopes were observed in postnatal lymphoid and nonhematopoietic tissues, with the class I and class II CD164 epitopes generally exhibiting differential and often reciprocal cellular distribution patterns. This is particularly striking in the colon, where infiltrating lymphoid cells are CD164 class I-positive but class II-negative, while epithelia are weakly CD164 class II-positive. Similarly, in certain lymphoid tissues, high endothelial venules and basal and subcapsular epithelia are CD164 class II-positive, while lymphoid cells are CD164 class I-positive. It therefore seems highly likely that these CD164 class I and II epitopes will mediate reciprocal homing functions in these tissue types.


Assuntos
Antígenos CD , Epitopos/imunologia , Células-Tronco Hematopoéticas/imunologia , Moléculas de Adesão de Célula Nervosa , Receptores de Superfície Celular/imunologia , Adulto , Antígenos CD34/imunologia , Antígeno CD146 , Endolina , Mapeamento de Epitopos , Feto/imunologia , Hematopoese , Humanos , Glicoproteínas de Membrana/imunologia , Especificidade de Órgãos
20.
J Cell Sci ; 113 ( Pt 5): 767-78, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10671367

RESUMO

We have developed an in vivo assay for progenitor cells of the human tracheobronchial epithelium relying on the transplantation of human prenatal respiratory tissues into severe combined immunodeficiency mice. Engrafted embryonic or fetal open tracheobronchial rudiments are rapidly closed at each end by a neoformed membrane that we named the operculum. After 2-4 weeks, differentiated human respiratory epithelium covers both the native airway matrix and the new operculum. Human epithelial cells dissociated from either emerging embryonic lung primordia or mature xenografts were seeded in host human airway grafts, of which native epithelium had been eliminated by several cycles of freezing and thawing. All grafts seeded with donor epithelial cells and implanted back into SCID mice recovered a surface mucociliary epithelium expressing expected markers and secreting mucus. Spontaneous epithelium regrowth was never observed in control unseeded, denuded grafts. In some experiments, donor epithelial cells and host denuded airway were sex-mismatched and the donor origin of newly formed epithelial structures was confirmed by sex chromosome detection. After two rounds of seeding and reimplantation, a normal epithelium was observed to line the 3rd generation operculum. These observations substantiate a functional assay for human candidate airway epithelium stem cells.


Assuntos
Células Epiteliais/fisiologia , Mucosa Respiratória/citologia , Mucosa Respiratória/crescimento & desenvolvimento , Células-Tronco/fisiologia , Animais , Brônquios/embriologia , Brônquios/fisiologia , Brônquios/transplante , Diferenciação Celular/genética , Células Epiteliais/transplante , Transplante de Tecido Fetal , Humanos , Camundongos , Camundongos SCID , Regeneração/genética , Mucosa Respiratória/fisiologia , Mucosa Respiratória/transplante , Transplante de Células-Tronco , Traqueia/embriologia , Traqueia/fisiologia , Traqueia/transplante , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA