Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Transfusion ; 51(12): 2656-63, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21599671

RESUMO

BACKGROUND: Hematopoietic stem cell transplantation is commonly used to treat several oncohematologic diseases. The autologous hematopoietic progenitor cells collected through apheresis (HPC-A) must be cryopreserved and stored before use in vivo. Cell processing that precedes cryopreservation of HPC-A includes volume reduction aimed at reducing the amount of dimethyl sulfoxide used, as well as storage space. STUDY DESIGN AND METHODS: The aim of our study was to assess the effectiveness of volume reduction performed with an automated closed system, namely, the Sepax S100 cell separation device (Biosafe SA). A total of 165 procedures were carried out on concentrates collected from 104 adult and pediatric patients. As a control group, 30 HPC-A units processed according to the standard method (i.e., centrifugation at a speed of 850 × g for 10 minutes, followed by manual plasma reduction) were evaluated. RESULTS: The volume reduction obtained was 59% (range, 20.54%-84.21%; standard deviation [SD], ± 12.19%), going from 236 mL (range, 100-443 mL; SD, ± 80.41 mL) to 97 mL (range, 33.00-263.00 mL; SD, ± 47.41 mL); recovery of nucleated cells was 90% (range, 64.84%-105.93%; SD, ± 8.76%), while that of CD34+ cells was 91% (range, 59.30%-119.37%; SD, ± 13.30%). These values did not differ from those obtained using the standard method. Automated processing required 20 minutes versus 40 minutes of manual processing. DISCUSSION: Our data demonstrate that volume reduction carried out with the Sepax S100 automated system was particularly effective; cell recovery was excellent and the time spent was short. Moreover, the closed system allows cell processing to be carried out in a contamination-controlled environment, in accordance with good manufacturing practice guidelines.


Assuntos
Preservação de Sangue , Separação Celular , Criopreservação , Células-Tronco Hematopoéticas/citologia , Transplante de Células-Tronco de Sangue Periférico , Adolescente , Adulto , Idoso , Preservação de Sangue/instrumentação , Preservação de Sangue/métodos , Separação Celular/instrumentação , Separação Celular/métodos , Criança , Pré-Escolar , Criopreservação/instrumentação , Criopreservação/métodos , Crioprotetores/farmacologia , Dimetil Sulfóxido/farmacologia , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Neoplasias/terapia , Transplante Homólogo
2.
Hum Genet ; 126(4): 511-20, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19484265

RESUMO

Deletion of chromosome 9p21 is a crucial event for the development of several cancers including acute lymphoblastic leukemia (ALL). Double strand breaks (DSBs) triggering 9p21 deletions in ALL have been reported to occur at a few defined sites by illegitimate action of the V(D)J recombination activating protein complex. We have cloned 23 breakpoint junctions for a total of 46 breakpoints in 17 childhood ALL (9 B- and 8 T-lineages) showing different size deletions at one or both homologous chromosomes 9 to investigate which particular sequences make the region susceptible to interstitial deletion. We found that half of 9p21 deletion breakpoints were mediated by ectopic V(D)J recombination mechanisms whereas the remaining half were associated to repeated sequences, including some with potential for non-B DNA structure formation. Other mechanisms, such as microhomology-mediated repair, that are common in other cancers, play only a very minor role in ALL. Nucleotide insertions at breakpoint junctions and microinversions flanking the breakpoints have been detected at 20/23 and 2/23 breakpoint junctions, respectively, both in the presence of recombination signal sequence (RSS)-like sequences and of other unspecific sequences. The majority of breakpoints were unique except for two cases, both T-ALL, showing identical deletions. Four of the 46 breakpoints coincide with those reported in other cases, thus confirming the presence of recurrent deletion hotspots. Among the six cases with heterozygous 9p deletions, we found that the remaining CDKN2A and CDKN2B alleles were hypermethylated at CpG islands.


Assuntos
Cromossomos Humanos Par 9/genética , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Deleção de Sequência/genética , Adolescente , Criança , Pré-Escolar , Quebra Cromossômica , Hibridização Genômica Comparativa , Ilhas de CpG , Metilação de DNA , Primers do DNA , Feminino , Heterozigoto , Humanos , Lactente , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas/genética
3.
Haematologica ; 94(12): 1649-60, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19773264

RESUMO

BACKGROUND: Mesenchymal stromal cells are employed in various different clinical settings in order to modulate immune response. However, relatively little is known about the mechanisms responsible for their immunomodulatory effects, which could be influenced by both the cell source and culture conditions. DESIGN AND METHODS: We tested the ability of a 5% platelet lysate-supplemented medium to support isolation and ex vivo expansion of mesenchymal stromal cells from full-term umbilical-cord blood. We also investigated the biological/functional properties of umbilical cord blood mesenchymal stromal cells, in comparison with platelet lysate-expanded bone marrow mesenchymal stromal cells. RESULTS: The success rate of isolation of mesenchymal stromal cells from umbilical cord blood was in the order of 20%. These cells exhibited typical morphology, immunophenotype and differentiation capacity. Although they have a low clonogenic efficiency, umbilical cord blood mesenchymal stromal cells may possess high proliferative potential. The genetic stability of these cells from umbilical cord blood was demonstrated by a normal molecular karyotype; in addition, these cells do not express hTERT and telomerase activity, do express p16(ink4a) protein and do not show anchorage-independent cell growth. Concerning alloantigen-specific immune responses, umbilical cord blood mesenchymal stromal cells were able to: (i) suppress T- and NK-lymphocyte proliferation, (ii) decrease cytotoxic activity and (iii) only slightly increase interleukin-10, while decreasing interferon-gamma secretion, in mixed lymphocyte culture supernatants. While an indoleamine 2,3-dioxygenase-specific inhibitor did not reverse mesenchymal stromal cell-induced suppressive effects, a prostaglandin E(2)-specific inhibitor hampered the suppressive effect of both umbilical cord blood- and bone marrow-mesenchymal stromal cells on alloantigen-induced cytotoxic activity. Mesenchymal stromal cells from both sources expressed HLA-G. CONCLUSIONS: Umbilical cord blood- and bone marrow-mesenchymal stromal cells may differ in terms of clonogenic efficiency, proliferative capacity and immunomodulatory properties; these differences may be relevant for clinical applications.


Assuntos
Plaquetas/química , Extratos Celulares/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Estromais/citologia , Antígenos CD/análise , Plaquetas/citologia , Western Blotting , Células da Medula Óssea/citologia , Diferenciação Celular/efeitos dos fármacos , Extratos Celulares/química , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Citotoxicidade Imunológica/imunologia , Feminino , Sangue Fetal/citologia , Citometria de Fluxo , Humanos , Imunofenotipagem , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Células-Tronco Mesenquimais/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/imunologia , Células Estromais/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Telomerase/genética
4.
Cytotherapy ; 11(7): 825-36, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19903096

RESUMO

BACKGROUND AIMS: Because of their capacity to modulate the immune response and promote tissue repair, mesenchymal stromal cells (MSC) represent a potential novel treatment for autoimmune/inflammatory diseases, including Crohn's disease (CD). The aim of the study was in vitro characterization of MSC from active CD patients for future clinical application. METHODS: MSC from the bone marrow (BM) of seven CD patients (median age 32 years) were expanded ex vivo in the presence of 5% platelet lysate; cells were investigated for clonogenic efficiency, proliferative capacity, morphology, immunophenotype, differentiation potential, genetic stability and ability to suppress in vitro proliferation of both autologous and allogeneic lymphocytes to polyclonal mitogens. Results were compared with those of BM MSC of four healthy donors (HD). RESULTS: MSC were successfully expanded from all patients. Colony-forming unit-fibroblast (CFU-F) frequency and proliferative capacity were comparable in CD and HD MSC. CD MSC showed typical spindle-shaped morphology and differentiated into osteoblasts, adipocytes and chondrocytes. Surface immunologic markers did not differ between CD and HD MSC, with the only exception of sizeable levels of HLA-DR at early culture passages [12-84% at passage (P)1] in the former. CD MSC ceased their growth at variable passages (from P8 to P25) and entered senescence without any change in morphology/proliferation rate. Array-comparative genomic hybridization demonstrated that CD MSC do not show imbalanced chromosomal rearrangements. Both CD and HD MSC inhibited in vitro proliferation of lymphocytes to mitogens. CONCLUSIONS: CD MSC show biologic characteristics similar to HD MSC and can be considered for anti-inflammatory and reparative cell therapy approaches in patients with refractory disease.


Assuntos
Doença de Crohn/terapia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Multipotentes/metabolismo , Adolescente , Adulto , Medula Óssea/patologia , Diferenciação Celular , Proliferação de Células , Feminino , Humanos , Imunofenotipagem , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/patologia , Pessoa de Meia-Idade , Células-Tronco Multipotentes/imunologia , Células-Tronco Multipotentes/patologia , Células-Tronco
5.
Cancer Res ; 67(19): 9142-9, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17909019

RESUMO

Significant improvement in the understanding of mesenchymal stem cell (MSC) biology has opened the way to their clinical use. However, concerns regarding the possibility that MSCs undergo malignant transformation have been raised. We investigated the susceptibility to transformation of human bone marrow (BM)-derived MSCs at different in vitro culture time points. MSCs were isolated from BM of 10 healthy donors and propagated in vitro until reaching either senescence or passage (P) 25. MSCs in the senescence phase were closely monitored for 8 to 12 weeks before interrupting the cultures. The genetic characterization of MSCs was investigated through array-comparative genomic hybridization (array-CGH), conventional karyotyping, and subtelomeric fluorescent in situ hybridization analysis both before and after prolonged culture. MSCs were tested for the expression of telomerase activity, human telomerase reverse transcriptase (hTERT) transcripts, and alternative lengthening of telomere (ALT) mechanism at different passages. A huge variability in terms of proliferative capacity and MSCs life span was noted between donors. In eight of 10 donors, MSCs displayed a progressive decrease in proliferative capacity until reaching senescence. In the remaining two MSC samples, the cultures were interrupted at P25 to pursue data analysis. Array-CGH and cytogenetic analyses showed that MSCs expanded in vitro did not show chromosomal abnormalities. Telomerase activity and hTERT transcripts were not expressed in any of the examined cultures and telomeres shortened during the culture period. ALT was not evidenced in the MSCs tested. BM-derived MSCs can be safely expanded in vitro and are not susceptible to malignant transformation, thus rendering these cells suitable for cell therapy approaches.


Assuntos
Células da Medula Óssea/patologia , Transformação Celular Neoplásica/patologia , Células-Tronco Mesenquimais/patologia , Telômero/genética , Células da Medula Óssea/fisiologia , Células da Medula Óssea/ultraestrutura , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/ultraestrutura , Células Cultivadas , Senescência Celular/fisiologia , Genes p53 , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Mesenquimais/ultraestrutura , Hibridização de Ácido Nucleico , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Telomerase/biossíntese , Telomerase/genética , Telômero/metabolismo , Fatores de Tempo
6.
Nephrol Dial Transplant ; 23(4): 1196-202, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18029377

RESUMO

BACKGROUND: Antibodies directed against alloantigens are implicated in the pathogenesis of several immune reactions complicating transplantation, including humoral rejection after solid organ transplantation. Mesenchymal stem cells (MSCs) have immunomodulatory capacity, since in vivo they may prolong skin graft survival in the animal model and can rescue patients with life-threatening graft-versus-host disease. METHODS: To investigate whether MSCs exert an inhibitory effect on antibody production during allostimulation, we stimulated peripheral blood mononuclear cells, obtained from healthy controls or sensitized patients undergoing dialysis for end-stage renal failure, in mixed lymphocyte culture (MLC), and evaluated immunoglobulin production either in the absence or in the presence of third-party allogeneic MSCs. We also evaluated the effect of MSCs on B-cell allostimulation performed adding to MLC a polyclonal stimulus delivered by an agonist anti-CD40 monoclonal antibody. RESULTS: We found that the addition of MSCs at the beginning of MLC considerably inhibited immunoglobulin production in standard MLC, irrespective of the MSC dose employed. Conversely, immunoglobulin secretion induced by direct CD40-CD40L binding was not significantly inhibited. Furthermore, we demonstrated, in one sensitized patient, that secretion of donor-specific anti-HLA class I antibodies detected both in baseline serum and in the supernatant of control MLC was inhibited by the addition of MSCs. Mechanistically, the addition of MSCs induced a striking decrease of IL-5 production in the cultures. CONCLUSIONS: Our findings suggest that third-party MSC are able to suppress allo-specific antibody production in vitro, and may therefore help overcome a positive cross-match in sensitized transplant recipients.


Assuntos
Antígenos CD19/imunologia , Linfócitos B/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Isoanticorpos/imunologia , Falência Renal Crônica/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/imunologia , Adulto , Antígenos CD40/imunologia , Células Cultivadas , Criança , Testes Imunológicos de Citotoxicidade , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulinas/biossíntese , Interleucinas/biossíntese , Isoantígenos/imunologia , Falência Renal Crônica/imunologia , Falência Renal Crônica/patologia , Células-Tronco Mesenquimais/citologia , Transplante Homólogo/imunologia
7.
N Engl J Med ; 351(14): 1419-24, 2004 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-15459303

RESUMO

A 27-year-old man with the autoimmune lymphoproliferative syndrome and a large-B-cell lymphoma had heterozygous mutations in the Fas and perforin (Prf1) genes. The Fas mutation was inherited from his healthy father and was also carried by his healthy brother, whereas the Prf1 mutation was inherited from his healthy mother. The combined effect of the two mutant genes may have contributed to the development of the autoimmune lymphoproliferative syndrome and lymphoma in this patient.


Assuntos
Doenças Autoimunes/genética , Linfoma de Células B/genética , Transtornos Linfoproliferativos/genética , Glicoproteínas de Membrana/genética , Mutação Puntual , Receptor fas/genética , Adulto , Feminino , Humanos , Linfoma Difuso de Grandes Células B/genética , Masculino , Linhagem , Perforina , Proteínas Citotóxicas Formadoras de Poros , Análise de Sequência de DNA
8.
Exp Hematol ; 34(7): 934-42, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16797421

RESUMO

OBJECTIVE: Multipotent mesenchymal stromal cells (MSCs) are endowed with multilineage differentiative potential and immunomodulatory properties. It is still a matter of debate whether donor MSCs have sustained engraftment potential in host bone marrow (BM) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). The aim of this study was to analyze the donor/recipient origin of MSCs in children receiving allogeneic either BM or cord blood (CB) transplantation. METHODS: Thirty-seven pediatric patients undergoing allo-HSCT for either a malignant or a nonmalignant disorder were enrolled in the study; 19 received CB and 18 BM transplantation. Results were compared with those obtained in 14 adults given BM transplantation for either malignant or nonmalignant disorders. MSCs were grown from BM aspirates obtained 1-17 and 2-192 months after allo-HSCT in pediatric and adult patients, respectively. MSC samples at the third-fourth passage were phenotypically characterized. Donor/recipient origin of MSCs was assessed by amelogenin assay and microsatellite analysis. RESULTS: MSCs could be grown from 30 of 37 children; at the third-fourth passage MSCs resulted positive (> or = 98%) for CD73, CD105, CD106, CD29, CD13, CD44 and negative (< or = 1%) for CD34, CD45, CD14. Mixed chimerism with donor cells was observed in 4 BM and 5 CB transplantation recipients, respectively; full recipient chimerism was detected in the remaining children. Full recipient MSC chimerism was observed also in all assessable (12/14) adult patients. CONCLUSIONS: BM of pediatric patients might be a more favorable milieu than that of adults for sustained engraftment of transplanted MSCs. MSCs able to engraft in the host can be transferred with cryopreserved CB units.


Assuntos
Transplante de Medula Óssea , Sangue Fetal/transplante , Mesoderma/citologia , Células Estromais/citologia , Adolescente , Adulto , Criança , Pré-Escolar , Humanos , Lactente
9.
Haematologica ; 90(4): 516-25, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15820948

RESUMO

BACKGROUND AND OBJECTIVES: Experimental evidence and preliminary clinical studies have demonstrated that human mesenchymal stem cells (MSC) have an important immune modulatory function in the setting of allogeneic hematopoietic stem cell (HSC) transplantation. We extended the evaluation of mechanisms responsible for the immune regulatory effect derived from the interaction of human MSC with cells involved in alloantigen-specific immune response in mixed lymphocyte culture (MLC). DESIGN AND METHODS: Dendritic cell (DC) differentiation, T- and natural killer (NK)-lymphocyte expansion, alloantigen-specific cytotoxic activity and differentiation of CD4+ T-cell subsets co-expressing CD25 and/or CTLA4 molecules were assessed, comparing the effect observed using third-party MSC with that obtained employing MSC autologous to the MLC responder. RESULTS: We found that human MSC strongly inhibit alloantigen-induced DC1 differentiation, down-regulate alloantigen-induced lymphocyte expansion, especially that of CD8+ T cells and of NK lymphocytes, decrease alloantigen-specific cytotoxic capacity mediated by either cytotoxic T lymphocytes or NK cells and favor the differentiation of CD4+ T-cell subsets co-expressing CD25 and/or CTLA4. More effective suppressive activity on MLC-induced T-cell activation was observed when MSC were third-party, rather than autologous, with respect to MLC-responder cells. INTERPRETATION AND CONCLUSIONS: Our results strongly suggest that MSC-mediated inhibition of alloantigen-induced DC1 differentiation and preferential activation of CD4+ CD25+ T-cell subsets with presumed regulatory activity represent important mechanisms contributing to the immunosuppressive activity of MSC. Collectively, these data provide immunological support for the use of MSC to prevent immune complications related to both HSC and solid organ transplantation and to the theory that MSC are universal suppressors of immune reactivity.


Assuntos
Linfócitos T CD4-Positivos/citologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Isoantígenos/imunologia , Células-Tronco Mesenquimais/citologia , Subpopulações de Linfócitos T/citologia , Formação de Anticorpos , Comunicação Celular , Diferenciação Celular , Epitopos , Citometria de Fluxo , Regulação da Expressão Gênica/genética , Humanos , Terapia de Imunossupressão , Teste de Cultura Mista de Linfócitos , Células-Tronco Mesenquimais/imunologia , Fenótipo
10.
Transplantation ; 78(8): 1229-32, 2004 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-15502726

RESUMO

Failure to mount or maintain a protective immune response may influence the development of polyomavirus BK (BKV)-associated nephropathy (PVAN). However, limited data are so far available on BKV-specific immunity after kidney transplantation. BKV-specific cellular immune response was retrospectively analyzed in kidney recipients with or without BKV infection/reactivation by measuring the frequency of interferon (IFN)-gamma-secreting cells in peripheral blood. Patients with BKV-active infection and good renal function (n=6) had a mean BKV-specific lymphocyte frequency 2 log lower than healthy controls and in the same range as BKV-seropositive recipients without active infection (n=7). Patients with PVAN (n=5) revealed undetectable levels of BKV-specific cells. However, two patients from the latter cohort treated with immunosuppression reduction showed the emergence of specific immunity, with IFN-gamma production in the same range as healthy controls. Our preliminary data suggest that lack of protective immunity toward BKV may favor the occurrence of BKV active infection and influence the progression to PVAN.


Assuntos
Vírus BK/imunologia , Epitopos , Imunidade Celular , Transplante de Rim/imunologia , Vírus BK/fisiologia , Humanos , Interferon gama/sangue , Rim/fisiopatologia , Nefropatias/imunologia , Nefropatias/virologia , Contagem de Linfócitos , Linfócitos/metabolismo , Infecções por Polyomavirus/sangue , Infecções por Polyomavirus/fisiopatologia , Período Pós-Operatório , Estudos Retrospectivos , Infecções Tumorais por Vírus/sangue , Infecções Tumorais por Vírus/fisiopatologia , Ativação Viral
11.
Pediatrics ; 129(1): e189-94, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22144701

RESUMO

Epstein-Barr virus (EBV)-related post-transplantation lymphoproliferative disorder (PTLD) is a life-threatening complication in patients given T-cell-depleted hematopoietic stem cell transplantation from an HLA-haploidentical relative (haplo-HSCT). We report the case of a child who developed severe EBV-related PTLD after haplo-HSCT from his mother. Despite receiving the anti-CD20 monoclonal antibody, the patient presented with intestinal obstruction due to huge abdominal lymphadenopathy, hematemesis, and nodulary pulmonary lesions. Histology showed that the lesions were due to CD20-/CD19+ large neoplastic B cells. The patient underwent double intestinal resection with partial abdominal lymphadenectomy and then received 3 monthly doses of donor-derived unmanipulated mononuclear cells. The initial dose of CD3+ cells was 3 10(5)/kg recipient body weight. The 2 additional doses consisted of 5 10(5) CD3+ cells/kg. No sign or symptom attributable to graft-versus-host disease was observed, and the patient completely cleared EBV-related lesions. The child was disease-free for 13 months after the first lymphocyte infusion. This case demonstrates that repeated infusions of controlled numbers of donor CD3+ cells cure EBV-related PTLD in haplo-HSCT without inducing graft-versus-host disease.


Assuntos
Infecções por Vírus Epstein-Barr/terapia , Antígenos HLA/análise , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Depleção Linfocítica , Transfusão de Linfócitos , Transtornos Linfoproliferativos/terapia , Anticorpos Monoclonais Murinos/uso terapêutico , Complexo CD3/análise , Pré-Escolar , Infecções por Vírus Epstein-Barr/complicações , Humanos , Imunossupressores/uso terapêutico , Linfócitos/imunologia , Transtornos Linfoproliferativos/etiologia , Transtornos Linfoproliferativos/virologia , Masculino , Rituximab , Linfócitos T , Transplante Homólogo
12.
Best Pract Res Clin Haematol ; 24(1): 73-81, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21396595

RESUMO

Mesenchymal stromal cells (MSCs) are adult multipotent cells that can be isolated from several human tissues. MSCs represent a novel and attractive tool in strategies of cellular therapy. For in vivo use, MSCs have to be ex vivo expanded in order to reach the numbers suitable for their clinical application. Despite being efficacious, the use of fetal calf serum for MSC ex vivo expansion for clinical purposes raises concerns related to immunization and transmission of zoonoses; the standardization of expansion methods, possibly devoid of animal components, such as those based on platelet lysate, are discussed in this paper. Moreover, this review focuses on the search of novel markers for the prospective identification/isolation of MSCs and on the potential risks connected with ex vivo expansion of MSCs, in particular that of their malignant transformation. Available tests to study the genetic stability of ex vivo expanded MSCs are also analyzed.


Assuntos
Proliferação de Células , Células-Tronco Mesenquimais/citologia , Células Estromais/citologia , Células da Medula Óssea , Técnicas de Cultura de Células/métodos , Separação Celular , Células Cultivadas , Meios de Cultura Livres de Soro , Humanos , Células-Tronco Multipotentes
13.
J Biomed Mater Res A ; 94(3): 790-9, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20336739

RESUMO

The use of stem cells in regenerative medicine is an appealing area of research that has received a great deal of interest in recent years. The population called human adipose tissue-derived stem cells (hASCs) share many of the characteristic of its counterpart of marrow including extensive proliferative potential and the ability to undergo multilineage differentiation along classical mesenchymal lineages: adipogenesis, chondrogenesis, osteogenesis, and myogenesis. The aim of this study was to evaluate with biochemical and morphological methods the adhesion and differentiation of hASCs grown on trabecular titanium scaffolds. The hASCs isolated from subcutaneous adipose tissue after digestion with collagenase were seeded on monolayer and on trabecular titanium scaffolds and incubated at 37 degrees C in 5% CO(2) with osteogenic medium or control medium.The results showed that hASCs were able to adhere to titanium scaffolds, to proliferate, to acquire an osteoblastic-like phenotype, and to produce a calcified extracellular matrix with protein, such as, decorin, fibronectin, osteocalcin, osteonectin, osteopontin, and type I collagen. These data suggest that this kind of scaffold/cells construct is effective to regenerate damaged tissue and to restore the function of bone tissue.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Osteoblastos/metabolismo , Células-Tronco/fisiologia , Alicerces Teciduais , Titânio/metabolismo , Tecido Adiposo/citologia , Ligas , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Células Cultivadas , Humanos , Teste de Materiais , Osteoblastos/citologia , Células-Tronco/citologia , Engenharia Tecidual/métodos , Titânio/química
14.
Diabetes ; 57(4): 1078-83, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18198357

RESUMO

OBJECTIVE: Perforin plays a key role in cell-mediated cytotoxicity. Mutations of its gene, PRF1, cause familial hemophagocytic lymphohistiocytosis but have also been associated with lymphomas and the autoimmune/lymphoproliferative syndrome. The aim of this work was to investigate the role of PRF1 variations in type 1 diabetes. RESEARCH DESIGN AND METHODS: We typed for the N252S and A91V variations in an initial population of 352 type 1 diabetic patients and 816 control subjects and a second population of 365 patients and 964 control subjects. Moreover, we sequenced the coding sequence and intron-exons boundaries in 200 patients and 300 control subjects. RESULTS: In both cohorts, allelic frequency of N252S was significantly higher in patients than in control subjects (combined cohorts: 1.5 vs. 0.4%; odds ratio 6.68 [95% CI 1.83-7.48]). Sequencing of the entire coding region detected one novel mutation in one patient, causing a P477A amino acid change not detected in 199 patients and 300 control subjects. Typing for HLA-DQA1 and DQB1 alleles showed that type 1 diabetes-predisposing DQ alpha/DQ beta heterodimers were less frequent in patients carrying N252S or P477A than in those carrying wild-type PRF1. We previously found that natural killer (NK) activity is not decreased in most N252S heterozygotes, but we detected one whose NK activity was normal at the age of 12 but strikingly low in early childhood. Here, we discovered that NK function was low in three heterozygotes in early childhood, one homozygous adult, and in the subject carrying P477A. CONCLUSIONS: These data suggest that N252S and possibly other PRF1 variations are susceptibility factors for type 1 diabetes development.


Assuntos
Diabetes Mellitus Tipo 1/genética , Variação Genética , Proteínas Citotóxicas Formadoras de Poros/genética , Adolescente , Adulto , Substituição de Aminoácidos , Estudos de Coortes , Primers do DNA , Diabetes Mellitus Tipo 1/imunologia , Feminino , Frequência do Gene , Predisposição Genética para Doença , Antígenos HLA-DQ/genética , Cadeias alfa de HLA-DQ , Cadeias beta de HLA-DQ , Humanos , Itália , Masculino , Perforina , Polimorfismo de Nucleotídeo Único , Valores de Referência
15.
Blood ; 110(7): 2764-7, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17638847

RESUMO

Haploidentical hematopoietic stem-cell transplantation (HSCT) is associated with an increased risk of graft failure. Adult bone marrow-derived mesenchymal stromal cells (MSCs) have been shown to support in vivo normal hematopoiesis and to display potent immune suppressive effects. We cotransplanted donor MSCs in 14 children undergoing transplantation of HLA-disparate CD34(+) cells from a relative. While we observed a graft failure rate of 15% in 47 historic controls, all patients given MSCs showed sustained hematopoietic engraftment without any adverse reaction. In particular, children given MSCs did not experience more infections compared with controls. These data suggest that MSCs, possibly thanks to their potent immunosuppressive effect on alloreactive host T lymphocytes escaping the preparative regimen, reduce the risk of graft failure in haploidentical HSC transplant recipients.


Assuntos
Técnicas de Cultura de Células/métodos , Rejeição de Enxerto/prevenção & controle , Transplante de Células-Tronco Hematopoéticas , Linfócitos/citologia , Linfócitos/imunologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Adolescente , Proliferação de Células , Criança , Pré-Escolar , Feminino , Seguimentos , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Humanos , Lactente , Masculino , Células-Tronco Mesenquimais/imunologia , Fatores de Risco
16.
Blood ; 108(9): 3079-84, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16720836

RESUMO

Mutations decreasing function of the Fas death receptor cause the autoimmune lymphoproliferative syndrome (ALPS) with autoimmune manifestations, spleen/lymph node enlargement, and expansion of CD4/CD8-negative T cells. Dianzani Autoimmune Lymphoproliferative Disease (DALD) is a variant lacking this expansion. Perforin is involved in cell-mediated cytotoxicity and its biallelic mutations cause familial hemophagocytic lymphohistiocytosis (HLH). We previously described an ALPS patient carrying heterozygous mutations of the Fas and perforin genes and suggested that they concurred in ALPS. This work extends the analysis to 14 ALPS, 28 DALD, and 816 controls, and detects an N252S amino acid substitution in 2 ALPS, and an A91V amino acid substitution in 6 DALD. N252S conferred an OR = 62.7 (P = .0016) for ALPS and A91V conferred an OR = 3 (P = .016) for DALD. Copresence of A91V and variations of the osteopontin gene previously associated with DALD conferred an OR = 17 (P = .0007) for DALD. In one N252S patient, NK activity was strikingly defective in early childhood, but became normal in late childhood. A91V patients displayed lower NK activity than controls. These data suggest that perforin variations are a susceptibility factor for ALPS/DALD development in subjects with defective Fas function and may influence disease expression.


Assuntos
Doenças Autoimunes/genética , Variação Genética , Transtornos Linfoproliferativos/genética , Glicoproteínas de Membrana/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Receptor fas/fisiologia , Substituição de Aminoácidos , Citotoxicidade Imunológica , Genótipo , Humanos , Células Matadoras Naturais/imunologia , Contagem de Linfócitos , Perforina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA