Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 45
Filtrer
1.
Stem Cell Res ; 49: 102035, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33221677

RÉSUMÉ

The development of induced pluripotent stem cells (iPSCs) by Shinya Yamanaka and colleagues in 2006 has led to a potential new paradigm in cellular therapeutics, including the possibility of producing patient-specific, disease-specific and immune matched allogeneic cell therapies. One can envisage two routes to immunologically compatible iPSC therapies: using genetic modification to generate a 'universal donor' with reduced expression of Human Leukocyte Antigens (HLA) and other immunological targets or developing a haplobank containing iPSC lines specifically selected to provide HLA matched products to large portions of the population. HLA matched lines can be stored in a designated physical or virtual global bank termed a 'haplobank'. The process of 'iPSC haplobanking' refers to the banking of iPSC cell lines, selected to be homozygous for different HLA haplotypes, from which therapeutic products can be derived and matched immunologically to patient populations. By matching iPSC and derived products to a patient's HLA class I and II molecules, one would hope to significantly reduce the risk of immune rejection and the use of immunosuppressive medication. Immunosuppressive drugs are used in several conditions (including autoimmune disease and in transplantation procedures) to reduce rejection of infused cells, or transplanted tissue and organs, due to major and minor histocompatibility differences between donor and recipient. Such regimens can lead to immune compromise and pathological consequences such as opportunistic infections or malignancies due to decreased cancer immune surveillance. In this article, we will discuss what is practically involved if one is developing and executing an iPSC haplobanking strategy.


Sujet(s)
Cellules souches pluripotentes induites , Banques de tissus , Lignée cellulaire , Antigènes HLA/génétique , Haplotypes , Humains , Donneurs de tissus
2.
Stem Cells ; 38(1): 67-79, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31621975

RÉSUMÉ

Cell types differentiated from induced pluripotent stem cells (iPSCs) are frequently arrested in their development program, more closely resembling a fetal rather than an adult phenotype, potentially limiting their utility for downstream clinical applications. The fetal phenotype of iPSC-derived dendritic cells (ipDCs) is evidenced by their low expression of MHC class II and costimulatory molecules, impaired secretion of IL-12, and poor responsiveness to conventional maturation stimuli, undermining their use for applications such as immune-oncology. Given that iPSCs display an epigenetic memory of the cell type from which they were originally derived, we investigated the feasibility of reprogramming adult DCs to pluripotency to determine the impact on the phenotype and function of ipDCs differentiated from them. Using murine bone marrow-derived DCs (bmDCs) as proof of principle, we show here that immature DCs are tractable candidates for reprogramming using non-integrating Sendai virus for the delivery of Oct4, Sox2, Klf4, and c-Myc transcription factors. Reprogramming efficiency of DCs was lower than mouse embryonic fibroblasts (MEFs) and highly dependent on their maturation status. Although control iPSCs derived from conventional MEFs yielded DCs that displayed a predictable fetal phenotype and impaired immunostimulatory capacity in vitro and in vivo, DCs differentiated from DC-derived iPSCs exhibited a surface phenotype, immunostimulatory capacity, and responsiveness to maturation stimuli indistinguishable from the source DCs, a phenotype that was retained for 15 passages of the parent iPSCs. Our results suggest that the epigenetic memory of iPSCs may be productively exploited for the generation of potently immunogenic DCs for immunotherapeutic applications.


Sujet(s)
Reprogrammation cellulaire/génétique , Cellules dendritiques/métabolisme , Immunothérapie/méthodes , Cellules souches pluripotentes induites/métabolisme , Animaux , Différenciation cellulaire , Humains , Facteur-4 de type Kruppel , Souris
3.
Trends Mol Med ; 25(11): 935-937, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31624020

RÉSUMÉ

Immune checkpoint inhibitors (ICIs) have revolutionised cancer immunotherapy but their success is wholly dependent on amplifying an existing immune response directed against the tumour. A recent study by Tsuchiya et al. suggests how the properties of induced pluripotent stem cells (iPSCs) may be exploited for the targeted delivery of interferon-α (IFNα) to elicit an appropriate response.


Sujet(s)
Immunothérapie , Interféron alpha , Thérapie moléculaire ciblée , Tumeurs/immunologie , Humains , Interféron alpha/métabolisme , Interféron alpha/pharmacologie , Tumeurs/thérapie , Cellules souches pluripotentes/immunologie , Cellules souches pluripotentes/métabolisme
4.
Biomed J ; 40(2): 80-93, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-28521905

RÉSUMÉ

The acquisition of self-perpetuating, immunological tolerance specific for graft alloantigens has long been described as the "holy grail" of clinical transplantation. By removing the need for life-long immunosuppression following engraftment, the adverse consequences of immunosuppressive regimens, including chronic infections and malignancy, may be avoided. Furthermore, autoimmune diseases and allergy are, by definition, driven by aberrant immunological responses to ordinarily innocuous antigens. The re-establishment of permanent tolerance towards instigating antigens may, therefore, provide a cure to these common diseases. Whilst various cell types exhibiting a tolerogenic phenotype have been proposed for such a task, tolerogenic dendritic cells (tol-DCs) are exquisitely adapted for antigen presentation and interact with many facets of the immune system: as such, they are attractive candidates for use in strategies for immune intervention. We review here our current understanding of tol-DC mediated induction and maintenance of immunological tolerance. Additionally, we discuss recent in vitro findings from animal models and clinical trials of tol-DC immunotherapy in the setting of transplantation, autoimmunity and allergy which highlight their promising therapeutic potential, and speculate how tol-DC therapy may be developed in the future.


Sujet(s)
Cellules dendritiques , Tolérance immunitaire/immunologie , Immunosuppresseurs/usage thérapeutique , Lymphocytes T régulateurs/immunologie , Animaux , Présentation d'antigène/immunologie , Cellules dendritiques/cytologie , Humains , Immunothérapie/méthodes
5.
Front Immunol ; 8: 1935, 2017.
Article de Anglais | MEDLINE | ID: mdl-29358940

RÉSUMÉ

The advent of induced pluripotent stem cells (iPSCs) has begun to revolutionize cell therapy by providing a convenient source of rare cell types not normally available from patients in sufficient numbers for therapeutic purposes. In particular, the development of protocols for the differentiation of populations of leukocytes as diverse as naïve T cells, macrophages, and natural killer cells provides opportunities for their scale-up and quality control prior to administration. One population of leukocytes whose therapeutic potential has yet to be explored is the subset of conventional dendritic cells (DCs) defined by their surface expression of CD141. While these cells stimulate cytotoxic T cells in response to inflammation through the cross-presentation of viral and tumor-associated antigens in an MHC class I-restricted manner, under steady-state conditions CD141+ DCs resident in interstitial tissues are focused on the maintenance of homeostasis through the induction of tolerance to local antigens. Here, we describe protocols for the directed differentiation of human iPSCs into a mixed population of CD11c+ DCs through the spontaneous formation of embryoid bodies and exposure to a cocktail of growth factors, the scheduled withdrawal of which serves to guide the process of differentiation. Furthermore, we describe the enrichment of DCs expressing CD141 through depletion of CD1c+ cells, thereby obtaining a population of "untouched" DCs unaffected by cross-linking of surface CD141. The resulting cells display characteristic phagocytic and endocytic capacity and acquire an immunostimulatory phenotype following exposure to inflammatory cytokines and toll-like receptor agonists. Nevertheless, under steady-state conditions, these cells share some of the tolerogenic properties of tissue-resident CD141+ DCs, which may be further reinforced by exposure to a range of pharmacological agents including interleukin-10, rapamycin, dexamethasone, and 1α,25-dihydoxyvitamin D3. Our protocols therefore provide access to a novel source of DCs analogous to the CD141+ subset under steady-state conditions in vivo and may, therefore, find utility in the treatment of a range of disease states requiring the establishment of immunological tolerance.

6.
Regen Med ; 11(8): 817-829, 2016 12.
Article de Anglais | MEDLINE | ID: mdl-27905261

RÉSUMÉ

Few topics in regenerative medicine have inspired such impassioned debate as the immunogenicity of cell types and tissues differentiated from pluripotent stem cells. While early predictions suggested that tissues derived from allogeneic sources may evade immune surveillance altogether, the pendulum has since swung to the opposite extreme, with reports that the ectopic expression of a few developmental antigens may prompt rejection, even of tissues differentiated from autologous cell lines. Here we review the evidence on which these contradictory claims are based in order to reach an objective assessment of the likely magnitude of the immunological challenges ahead. Furthermore, we discuss how the inherent properties of pluripotent stem cells may inform strategies for reducing the impact of immunogenicity on the future ambitions of regenerative medicine.


Sujet(s)
Différenciation cellulaire/immunologie , Immunité cellulaire/immunologie , Cellules souches pluripotentes/immunologie , Médecine régénérative , Animaux , Humains , Cellules souches pluripotentes/cytologie
8.
Regen Med ; 10(3): 275-86, 2015.
Article de Anglais | MEDLINE | ID: mdl-25933237

RÉSUMÉ

As the fulcrum on which the balance between the opposing forces of tolerance and immunity has been shown to pivot, dendritic cells (DC) hold significant promise for immune intervention in a variety of disease states. Here we discuss how the directed differentiation of human pluripotent stem cells may address many of the current obstacles to the use of monocyte-derived DC in immunotherapy, providing a novel source of previously inaccessible DC subsets and opportunities for their scale-up, quality control and genetic modification. Indeed, given that it is the immunological legacy DC leave behind that is of therapeutic value, rather than their persistence per se, we propose that immunotherapy should serve as an early target for the clinical application of pluripotent stem cells.


Sujet(s)
Cellules dendritiques/immunologie , Tolérance immunitaire , Immunothérapie/méthodes , Monocytes/immunologie , Cellules souches pluripotentes/immunologie , Humains
9.
Nanomedicine ; 11(4): 879-83, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25659648

RÉSUMÉ

Extracellular vesicles (EVs) are natural nanoparticles that mediate intercellular transfer of RNA and proteins and are of great medical interest; serving as novel biomarkers and potential therapeutic agents. However, there is little consensus on the most appropriate method to isolate high-yield and high-purity EVs from various biological fluids. Here, we describe a systematic comparison between two protocols for EV purification: ultrafiltration with subsequent liquid chromatography (UF-LC) and differential ultracentrifugation (UC). A significantly higher EV yield resulted from UF-LC as compared to UC, without affecting vesicle protein composition. Importantly, we provide novel evidence that, in contrast to UC-purified EVs, the biophysical properties of UF-LC-purified EVs are preserved, leading to a different in vivo biodistribution, with less accumulation in lungs. Finally, we show that UF-LC is scalable and adaptable for EV isolation from complex media types such as stem cell media, which is of huge significance for future clinical applications involving EVs. FROM THE CLINICAL EDITOR: Recent evidence suggests extracellular vesicles (EVs) as another route of cellular communication. These EVs may be utilized for future therapeutics. In this article, the authors compared ultrafiltration with size-exclusion liquid chromatography (UF-LC) and ultra-centrifugation (UC) for EV recovery.


Sujet(s)
Microparticules membranaires/composition chimique , Microparticules membranaires/ultrastructure , Chromatographie sur gel , Cellules HEK293 , Humains , Ultrafiltration
10.
Cell Reprogram ; 17(1): 7-18, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25549177

RÉSUMÉ

In spite of considerable interest in the field, reprogramming induced pluripotent stem cells (iPSCs) directly from cancer cells has encountered considerable challenges, including the extremely low reprogramming efficiency and instability of cancer-derived iPSCs (C-iPSCs). In this study, we aimed to identify the main obstacles that limit cancer cell reprogramming. Through a detailed multidimensional kinetic optimization, a highly optimized protocol is established for reprogramming C-iPSCs using nonviral plasmid vectors. We demonstrated how the initial cancer cell density seeded could be the most critical factor ultimately affecting C-iPSCs reprogramming. We have consistently achieved an unprecedented high C-iPSC reprogramming efficiency, establishing stable colonies with typical iPSC morphology, up to 50% of which express the iPSC phenotypic (Oct3/4, Sox2, Nanog) and enzymatic (alkaline phosphatase) markers. Furthermore, established C-iPSC lines were shown to be capable of forming teratomas in vivo, containing cell types and tissues from each of the embryonic germ layers, fully consistent with their acquisition of pluripotency. This protocol was tested and confirmed in two completely unrelated human lung adenocarcinoma (A549) and mouse melanoma (B16f10) cancer cell lines and thus offers a potentially valuable method for generating effectively virus-free C-iPSCs for future applications.


Sujet(s)
Reprogrammation cellulaire , Vecteurs génétiques , Cellules souches pluripotentes induites/cytologie , Plasmides/génétique , Transfection/méthodes , Animaux , Techniques de culture cellulaire , Différenciation cellulaire , Lignée cellulaire tumorale , Cellules cultivées , Humains , Souris , Tératome/anatomopathologie
11.
Methods Mol Biol ; 1029: 1-16, 2013.
Article de Anglais | MEDLINE | ID: mdl-23756938

RÉSUMÉ

Immune privilege provides protection to vital tissues or cells of the body when foreign antigens are introduced into these sites. The modern concept of relative immune privilege applies to a variety of tissues and anatomical structures, including the hair follicles and mucosal surfaces. Even sites of chronic inflammation and developing tumors may acquire immune privilege by recruiting immunoregulatory effector cells. Adult stem cells are no exception. For their importance and vitality, many adult stem cell populations are believed to be immune privileged. A preimplantation-stage embryo that derives from a totipotent stem cell (i.e., a fertilized oocyte) must be protected from maternal allo-rejection for successful implantation and development to occur. Embryonic stem cells, laboratory-derived cell lines of preimplantation blastocyst-origin, may, therefore, retain some of the immunological properties of the developing embryo. However, embryonic stem cells and their differentiated tissue derivatives transplanted into a recipient do not necessarily have an ability to subvert immune responses to the extent required to exploit their pluripotency for regenerative medicine. In this review, an extended definition of immune privilege is developed and the capacity of adult and embryonic stem cells to display both relative and acquired immune privilege is discussed. Furthermore, we explore how these intrinsic properties of stem cells may one day be harnessed for therapeutic gain.


Sujet(s)
Cellules souches/cytologie , Cellules souches/immunologie , Cellules souches adultes/cytologie , Cellules souches adultes/immunologie , Cellules souches embryonnaires/cytologie , Cellules souches embryonnaires/immunologie , Humains , Modèles biologiques , Médecine régénérative
12.
Expert Rev Clin Immunol ; 8(6): 547-55, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22992149

RÉSUMÉ

Dendritic cells (DCs) possess the specialized potential to present exogenously derived antigen to cytotoxic T lymphocytes to elicit an immune response. This process, termed cross presentation, is crucial in the generation of immune response to viruses, tumors and in autoimmune disease. The ability of DCs to cross-present exogenous antigen to cytotoxic T lymphocytes makes them an attractive target for exploitation in immunotherapy. In recent years, significant advances have been made in understanding the mechanism of cross-presentation and the DC subsets involved. The recent discovery of the human cross presenting DC has given this field a new lease of life. In this report, the authors provide an overview of cross-presentation of antigen by DCs, focusing on the current understanding of the molecular mechanisms of the process. The authors also discuss the DC subsets involved in cross presentation and its role in health and disease.


Sujet(s)
Présentation d'antigène/immunologie , Cross-priming/immunologie , Cellules dendritiques/immunologie , Humains
13.
J Biomed Biotechnol ; 2012: 172420, 2012.
Article de Anglais | MEDLINE | ID: mdl-22505805

RÉSUMÉ

While human embryonic stem cells (hESCs) may one day facilitate the treatment of degenerative diseases requiring cell replacement therapy, the success of regenerative medicine is predicated on overcoming the rejection of replacement tissues. Given the role played by dendritic cells (DCs) in the establishment of immunological tolerance, we have proposed that DC, rendered tolerogenic during their differentiation from hESC, might predispose recipients to accept replacement tissues. As a first step towards this goal, we demonstrate that DC differentiated from H1 hESCs (H1-DCs) are particularly responsive to the immunosuppressive agent rapamycin compared to monocyte-derived DC (moDC). While rapamycin had only modest impact on the phenotype and function of moDC, H1-DC failed to upregulate CD40 upon maturation and displayed reduced immunostimulatory capacity. Furthermore, coculture of naïve allogeneic T cells with rapamycin-treated H1-DC promoted an increased appearance of CD25(hi) Foxp3+ regulatory T cells, compared to moDC. Our findings suggest that conditioning of hESC-derived DC with rapamycin favours a tolerogenic phenotype.


Sujet(s)
Cellules dendritiques/cytologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/cytologie , Immunosuppresseurs/pharmacologie , Sirolimus/pharmacologie , Lymphocytes T CD4+/immunologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Cellules cultivées , Cellules dendritiques/immunologie , Humains , Tolérance immunitaire/effets des médicaments et des substances chimiques , Phénotype
14.
Stem Cells Dev ; 21(10): 1688-700, 2012 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-21933027

RÉSUMÉ

The derivation of pluripotent embryonic stem cells (ESCs) from a variety of genetic backgrounds remains a desirable objective in the generation of mice functionally deficient in genes of interest and the modeling of human disease. Nevertheless, disparity in the ease with which different strains of mice yield ESC lines has long been acknowledged. Indeed, the generation of bona fide ESCs from the non obese diabetic (NOD) mouse, a well-characterized model of human type I diabetes, has historically proved especially difficult to achieve. Here, we report the development of protocols for the derivation of novel ESC lines from C57Bl/6 mice based on the combined use of high concentrations of leukemia inhibitory factor and serum-replacement, which is equally applicable to fresh and cryo-preserved embryos. Further, we demonstrate the success of this approach using Balb/K and CBA/Ca mice, widely considered to be refractory strains. CBA/Ca ESCs contributed to the somatic germ layers of chimeras and displayed a very high competence at germline transmission. Importantly, we were able to use the same protocol for the derivation of ESC lines from nonpermissive NOD mice. These ESCs displayed a normal karyotype that was robustly stable during long-term culture, were capable of forming teratomas in vivo and germline competent chimeras after injection into recipient blastocysts. Further, these novel ESC lines efficiently formed embryoid bodies in vitro and could be directed in their differentiation along the dendritic cell lineage, thus illustrating their potential application to the generation of cell types of relevance to the pathogenesis of type I diabetes.


Sujet(s)
Techniques de culture cellulaire , Corps embryoïdes/physiologie , Animaux , Blastocyste/cytologie , Différenciation cellulaire , Cellules cultivées , Chimère , Techniques de coculture , Cellules dendritiques/cytologie , Diabète de type 1/anatomopathologie , Techniques de culture d'embryons , Corps embryoïdes/transplantation , Cellules souches embryonnaires/physiologie , Femelle , Analyse de profil d'expression de gènes , Instabilité du génome , Caryotype , Test de culture lymphocytaire mixte , Mâle , Souris , Souris de lignée C57BL , Souris de lignée CBA , Souris de lignée NOD
15.
PLoS One ; 7(12): e52232, 2012.
Article de Anglais | MEDLINE | ID: mdl-23284947

RÉSUMÉ

Using an in vitro differentiation protocol we isolated cells with the properties of dendritic cells (DCs) from immunologically refractive pluripotent murine embryonic stem cells (ESCs). These ES-derived dendritic cells (ESDCs) expressed cytokines and were able to present antigen to a T cell line. Infection of ESDCs with Salmonella Typhimurium stimulated the expression of immune cell markers and thousands of murine genes, many associated with the immune response. Consequently, this system provides a novel in vitro model, amenable to genetic modification, for monitoring host/pathogen interactions.


Sujet(s)
Cellules dendritiques/cytologie , Cellules dendritiques/immunologie , Cellules souches embryonnaires/cytologie , Salmonella typhimurium/immunologie , Salmonella typhimurium/physiologie , Animaux , Lignée cellulaire , Cellules cultivées , Cytométrie en flux , Souris , Microscopie confocale , Microscopie électronique , Séquençage par oligonucléotides en batterie , Réaction de polymérisation en chaine en temps réel
16.
Methods Mol Biol ; 767: 449-61, 2011.
Article de Anglais | MEDLINE | ID: mdl-21822895

RÉSUMÉ

Improving our understanding of the interactions between human dendritic cells (DCs) and T cells may contribute to the development of therapeutic strategies for a variety of immune-mediated disorders. The possibility of using DCs themselves as tools to manipulate immune responses opens even greater therapeutic avenues. Current methods of generating human DCs are both inadequate and susceptible to high levels of variability between individuals. DCs differentiated from human embryonic stem cells (hESCs) could provide a more reliable, consistent solution. DCs have now successfully been differentiated from hESCs and more recently this has been repeated using protocols that avoid the inclusion of animal products, an important modification for clinical use. We have developed a novel method for the generation of DCs from hESCs in the absence of animal products that does not necessitate a separate embryoid body (EB) generation step. The technique involves the use of four growth factors and their successive removal from culture, resulting in accumulation of DCs with phenotypic, morphological, and immunostimulatory properties comparable to those of classical human monocyte-derived DCs. In addition to the application of hESC-derived DCs in basic research and novel approaches to cancer immunotherapy, they may also play a central role in the field of regenerative medicine. Tolerogenic DCs differentiated from hESCs may be used to persuade the immune system of the recipients of cell replacement therapy to tolerate allogeneic tissues differentiated from the same hESC line. Such an approach may help to address the immunological barriers that threaten to derail the clinical application of hESCs.


Sujet(s)
Techniques de culture cellulaire/méthodes , Différenciation cellulaire , Cellules dendritiques/cytologie , Cellules souches embryonnaires/cytologie , Numération cellulaire , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Collagène/pharmacologie , Test clonogénique , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/métabolisme , Association médicamenteuse , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/métabolisme , Fibroblastes/cytologie , Fibroblastes/effets des médicaments et des substances chimiques , Humains , Laminine/pharmacologie , Monocytes/cytologie , Monocytes/effets des médicaments et des substances chimiques , Protéoglycanes/pharmacologie
17.
Curr Opin Organ Transplant ; 16(4): 372-8, 2011 Aug.
Article de Anglais | MEDLINE | ID: mdl-21666481

RÉSUMÉ

PURPOSE OF REVIEW: Although long-term immune suppression remains the intervention of choice for the treatment of allograft rejection, transplantation tolerance would achieve graft survival with fewer inherent risks. Although the use of dendritic cells for the induction of tolerance might confer antigen specificity, factors determining the balance between tolerogenicity and immunogenicity remain uncertain, as does the stability of the functional phenotype. Here, we review recent studies suggesting that pharmacological agents may profoundly influence this delicate balance and outline the insights they provide into parameters that contribute to the tolerogenic state. RECENT FINDINGS: Recent findings have revealed that the inhibition of dendritic cell maturation by pharmacological intervention is not a prerequisite for the acquisition of tolerogenicity, but that susceptibility to a tolerogenic phenotype may vary between dendritic cell subsets and depend on the nature of maturation stimuli to which the cells are exposed. Furthermore, such studies have highlighted the degree to which the maintenance of tolerogenicity is influenced by local environmental factors, such as the cytokine milieu. SUMMARY: Although the rational design of tolerogenic dendritic cells for modulating the outcome of organ transplantation remains ambitious, the use of pharmacological agents to influence their functional phenotype continues to illuminate the basic biology of this critical cell type.


Sujet(s)
Cellules dendritiques/effets des médicaments et des substances chimiques , Rejet du greffon/prévention et contrôle , Survie du greffon/effets des médicaments et des substances chimiques , Transplantation d'organe , Tolérance à la transplantation/effets des médicaments et des substances chimiques , Animaux , Cytokines/métabolisme , Cellules dendritiques/immunologie , Rejet du greffon/immunologie , Humains , Phénotype , Transduction du signal/effets des médicaments et des substances chimiques , Récepteurs de type Toll/effets des médicaments et des substances chimiques , Récepteurs de type Toll/métabolisme , Résultat thérapeutique
18.
Blood ; 117(15): 4008-11, 2011 Apr 14.
Article de Anglais | MEDLINE | ID: mdl-21343609

RÉSUMÉ

Induced pluripotent stem (iPS) cells offer a unique potential for understanding the molecular basis of disease and development. Here we have generated several human iPS cell lines, and we describe their pluripotent phenotype and ability to differentiate into erythroid cells, monocytes, and endothelial cells. More significantly, however, when these iPS cells were differentiated under conditions that promote lympho-hematopoiesis from human embryonic stem cells, we observed the formation of pre-B cells. These cells were CD45(+)CD19(+)CD10(+) and were positive for transcripts Pax5, IL7αR, λ-like, and VpreB receptor. Although they were negative for surface IgM and CD5 expression, iPS-derived CD45(+)CD19(+) cells also exhibited multiple genomic D-J(H) rearrangements, which supports a pre-B-cell identity. We therefore have been able to demonstrate, for the first time, that human iPS cells are able to undergo hematopoiesis that contributes to the B-cell lymphoid lineage.


Sujet(s)
Lymphocytes B/cytologie , Lymphopoïèse/physiologie , Cellules souches pluripotentes/cytologie , Précurseurs lymphoïdes B/cytologie , Adulte , Antigènes CD19/métabolisme , Lymphocytes B/physiologie , Lignée cellulaire , Lignage cellulaire/immunologie , Humains , Pseudochaines légères des immunoglobulines/génétique , Immunophénotypage , Antigènes CD45/métabolisme , Néprilysine/métabolisme , Protéine activatrice spécifique des lymphocytes B/génétique , Cellules souches pluripotentes/physiologie , Précurseurs lymphoïdes B/physiologie , Récepteurs à l'interleukine-7/génétique
19.
Nat Rev Immunol ; 10(12): 868-75, 2010 12.
Article de Anglais | MEDLINE | ID: mdl-21107347

RÉSUMÉ

Few advances have been so widely acclaimed in biology as the seminal demonstration that adult somatic cells can be induced to acquire the phenotype and differentiation potential of embryonic stem cells. The capacity to produce patient-specific stem cells that are truly pluripotent has raised prospects for the treatment of many degenerative diseases through replacement of the affected cell types. In the race to the clinic, however, questions surrounding the potential immunogenicity of such cells have been largely overlooked. Here, I explore the extent of the challenges ahead and suggest that the induction of tolerance to such cells will be crucial to the future success of induced pluripotency.


Sujet(s)
Transplantation cellulaire , Cellules souches embryonnaires/immunologie , Cellules souches pluripotentes induites/immunologie , Tolérance à la transplantation , Adulte , Animaux , Humains , Souris , Rats , Facteurs de transcription/génétique , Transduction génétique
20.
Stem Cells ; 28(10): 1905-14, 2010 Oct.
Article de Anglais | MEDLINE | ID: mdl-20737577

RÉSUMÉ

We have previously reported that ESC-derived tissues are subject to some level of immune privilege, which might facilitate induction of immune tolerance. Herein, we further demonstrate that fully allogeneic ESC-derived tissues are accepted with a regimen of coreceptor blockade even in recipients known to be relatively resistant to such a tolerizing protocol. Moreover, ESC-derived tissues could be spontaneously accepted across a class I major histocompatibility complex disparity. We further show that CD4(+)FoxP3(+) regulatory T cells (Treg) appear to be essential for this natural "privileged" state as their ablation with an anti-CD25 mAb results in rejection of ESC-derived tissue. This same treatment exposes activation of macrophages and effector CD8(+) T cells, suggesting that these cells are subject to regulatory T cell control. Thus, spontaneous acceptance of ESC-derived tissues mimics the acquired immune privilege induced by coreceptor blockade and is determined by Treg-mediated suppression.


Sujet(s)
Cellules souches embryonnaires/cytologie , Complexe majeur d'histocompatibilité/immunologie , Lymphocytes T régulateurs/immunologie , Tolérance à la transplantation/immunologie , Animaux , Lymphocytes T CD8+/immunologie , Lignée cellulaire , Femelle , Immunohistochimie , Mâle , Souris , Souris de lignée C57BL , Réaction de polymérisation en chaîne , Lymphocytes T régulateurs/métabolisme , Transplantation homologue/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...