Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Crit Rev Oncol Hematol ; 185: 103956, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36893946

RESUMEN

Neuroblastoma represents a relatively common childhood tumor that imposes therapeutic difficulties. High risk neuroblastoma patients have poor prognosis, display limited response to radiochemotherapy and may be treated by hematopoietic cell transplantation. Allogeneic and haploidentical transplants have the distinct advantage of reinstitution of immune surveillance, reinforced by antigenic barriers. The key factors favorable to ignition of potent anti-tumor reactions are transition to adaptive immunity, recovery from lymphopenia and removal of inhibitory signals that inactivate immune cells at the local and systemic levels. Post-transplant immunomodulation may further foster anti-tumor reactivity, with positive but transient impact of infusions of lymphocytes and natural killer cells both from the donor, the recipient or third party. The most promising approaches include introduction of antigen-presenting cells in early post-transplant stages and neutralization of inhibitory signals. Further studies will likely shed light on the nature and actions of suppressor factors within tumor stroma and at the systemic level.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Neuroblastoma , Humanos , Niño , Trasplante Homólogo , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Inmunoterapia , Neuroblastoma/terapia , Neuroblastoma/patología
2.
Blood Adv ; 7(10): 2181-2195, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-36780582

RESUMEN

Alloreactive T-effector cells (Teffs) are the major culprit of acute graft-versus-host disease (aGVHD) associated with hematopoietic stem cell transplantation. Ex vivo nonspecific depletion of T cells from the donor graft impedes stem cell engraftment and posttransplant immune reconstitution. Teffs upregulate Fas after activation and undergo Fas ligand (FasL)-mediated restimulation-induced cell death (RICD), an important mechanism of immune homeostasis. We targeted RICD as a means to eliminate host-reactive Teffs in vivo for the prevention of aGVHD. A novel form of FasL protein chimeric with streptavidin (SA-FasL) was transiently displayed on the surface of biotinylated lymphocytes, taking advantage of the high-affinity interaction between biotin and streptavidin. SA-FasL-engineered mouse and human T cells underwent apoptosis after activation in response to alloantigens in vitro and in vivo. SA-FasL on splenocytes was effective in preventing aGVHD in >70% of lethally irradiated haploidentical mouse recipients after cotransplantation with bone marrow cells, whereas all controls that underwent transplantation with nonengineered splenocytes developed aGVHD. Prevention of aGVHD was associated with an increased ratio of CD4+CD25+FoxP3+ T regulatory (Tregs) to Teffs and significantly reduced transcripts for proinflammatory cytokines in the lymphoid organs and target tissues. Depletion of Tregs from the donor graft abrogated the protection conferred by SA-FasL. This approach was also effective in a xenogeneic aGVHD setting where SA-FasL-engineered human PBMCs were transplanted into NSG mice. Direct display of SA-FasL protein on donor cells as an effective means of eliminating alloreactive Teffs in the host represents a practical approach with significant translation potential for the prevention of aGVHD.


Asunto(s)
Enfermedad Injerto contra Huésped , Ratones , Humanos , Animales , Proteína Ligando Fas , Estreptavidina , Enfermedad Injerto contra Huésped/prevención & control , Linfocitos T , Linfocitos
3.
Am J Transplant ; 20(5): 1285-1295, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31850658

RESUMEN

We have previously shown that pancreatic islets engineered to transiently display a modified form of FasL protein (SA-FasL) on their surface survive indefinitely in allogeneic recipients without a need for chronic immunosuppression. Mechanisms that confer long-term protection to allograft are yet to be elucidated. We herein demonstrated that immune protection evolves in two distinct phases; induction and maintenance. SA-FasL-engineered allogeneic islets survived indefinitely and conferred protection to a second set of donor-matched, but not third-party, unmanipulated islet grafts simultaneously transplanted under the contralateral kidney capsule. Protection at the induction phase involved a reduction in the frequency of proliferating alloreactive T cells in the graft-draining lymph nodes, and required phagocytes and TGF-ß. At the maintenance phase, immune protection evolved into graft site-restricted immune privilege as the destruction of long-surviving SA-FasL-islet grafts by streptozotocin followed by the transplantation of a second set of unmanipulated islet grafts into the same site from the donor, but not third party, resulted in indefinite survival. The induced immune privilege required both CD4+ CD25+ Foxp3+ Treg cells and persistent presence of donor antigens. Engineering cell and tissue surfaces with SA-FasL protein provides a practical, efficient, and safe means of localized immunomodulation with important implications for autoimmunity and transplantation.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Proteína Ligando Fas , Supervivencia de Injerto , Privilegio Inmunológico , Tolerancia Inmunológica
5.
Stem Cells Transl Med ; 6(3): 700-712, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28186688

RESUMEN

Hematopoietic chimerism is one of the effective approaches to induce tolerance to donor-derived tissue and organ grafts without administration of life-long immunosuppressive therapy. Although experimental efforts to develop such regimens have been ongoing for decades, substantial cumulative toxicity of combined hematopoietic and tissue transplants precludes wide clinical implementation. Tolerance is an active immunological process that includes both peripheral and central mechanisms of mutual education of coresident donor and host immune systems. The major stages include sequential suppression of early alloreactivity, establishment of hematopoietic chimerism and suppressor cells that sustain the state of tolerance, with significant mechanistic and temporal overlap along the tolerization process. Efforts to devise less toxic transplant strategies by reduction of preparatory conditioning focus on modulation rather than deletion of residual host immunity and early reinstitution of regulatory subsets at the central and peripheral levels. Stem Cells Translational Medicine 2017;6:700-712.


Asunto(s)
Quimerismo , Hematopoyesis/inmunología , Tolerancia Inmunológica , Animales , Rechazo de Injerto/inmunología , Humanos
6.
Clin Rev Allergy Immunol ; 52(3): 460-472, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27677500

RESUMEN

As physiological pathways of intercellular communication produced by all cells, cytokines are involved in the pathogenesis of inflammatory insulitis as well as pivotal mediators of immune homeostasis. Proinflammatory cytokines including interleukins, interferons, transforming growth factor-ß, tumor necrosis factor-α, and nitric oxide promote destructive insulitis in type 1 diabetes through amplification of the autoimmune reaction, direct toxicity to ß-cells, and sensitization of islets to apoptosis. The concept that neutralization of cytokines may be of therapeutic benefit has been tested in few clinical studies, which fell short of inducing sustained remission or achieving disease arrest. Therapeutic failure is explained by the redundant activities of individual cytokines and their combinations, which are rather dispensable in the process of destructive insulitis because other cytolytic pathways efficiently compensate their deficiency. Proinflammatory cytokines are less redundant in regulation of the inflammatory reaction, displaying protective effects through restriction of effector cell activity, reinforcement of suppressor cell function, and participation in islet recovery from injury. Our analysis suggests that the role of cytokines in immune homeostasis overrides their contribution to ß-cell death and may be used as potent immunomodulatory agents for therapeutic purposes rather than neutralized.


Asunto(s)
Anticuerpos Neutralizantes/uso terapéutico , Autoinmunidad , Diabetes Mellitus Tipo 1/terapia , Inmunoterapia/métodos , Células Secretoras de Insulina/patología , Apoptosis , Ensayos Clínicos como Asunto , Citocinas/inmunología , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Homeostasis , Humanos , Mediadores de Inflamación/metabolismo , Insuficiencia del Tratamiento
7.
Immunology ; 147(4): 377-88, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26749404

RESUMEN

Immune imbalance in autoimmune disorders such as type 1 diabetes may originate from aberrant activities of effector cells or dysfunction of suppressor cells. All possible defective mechanisms have been proposed for diabetes-prone species: (i) quantitative dominance of diabetogenic cells and decreased numbers of regulatory T cells, (ii) excessive aggression of effectors and defective function of suppressors, (iii) perturbed interaction between effector and suppressor cells, and (iv) variations in sensitivity to negative regulation. The experimental evidence available to date presents conflicting information on these mechanisms, with identification of perturbed equilibrium on the one hand and negation of critical role of each mechanism in propagation of diabetic autoimmunity on the other hand. In our analysis, there is no evidence that inherent abnormalities in numbers and function of effector and suppressor T cells are responsible for the immune imbalance responsible for propagation of type 1 diabetes as a chronic inflammatory process. Possibly, the experimental tools for investigation of these features of immune activity are still underdeveloped and lack sufficient resolution, in the presence of the extensive biological viability and functional versatility of effector and suppressor elements.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Ratones Endogámicos NOD , Animales , Apoptosis/inmunología , Comunicación Celular/inmunología , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1/metabolismo , Progresión de la Enfermedad , Inflamación/inmunología , Inflamación/metabolismo , Activación de Linfocitos/inmunología , Ratones , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
8.
Immunol Res ; 64(2): 360-8, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26639356

RESUMEN

The mechanisms of autoimmune reactivity onset in type 1 diabetes (T1D) remain elusive despite extensive experimentation and discussion. We reconsider several key aspects of the early stages of autoimmunity at four levels: islets, pancreatic lymph nodes, thymic function and peripheral immune homeostasis. Antigen presentation is the islets and has the capacity to provoke immune sensitization, either in the process of physiological neonatal ß cell apoptosis or as a consequence of cytolytic activity of self-reactive thymocytes that escaped negative regulation. Diabetogenic effectors are efficiently expanded in both the islets and the lymph nodes under conditions of empty lymphoid niches during a period of time coinciding with a synchronized wave of ß cell apoptosis surrounding weaning. A major drive of effector cell activation and expansion is inherent peripheral lymphopenia characteristic of neonates, though it remains unclear when is autoimmunity triggered in subjects displaying hyperglycemia in late adolescence. Our analysis suggests that T1D evolves through coordinated activity of multiple physiological mechanisms of stimulation within specific characteristics of the neonate immune system.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Animales , Apoptosis , Autoantígenos/inmunología , Susceptibilidad a Enfermedades , Homeostasis , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Vigilancia Inmunológica , Inflamación/inmunología , Islotes Pancreáticos/citología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Activación de Linfocitos , Linfocitos T/inmunología , Linfocitos T/metabolismo
9.
Immunol Res ; 64(1): 36-43, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26482052

RESUMEN

Two competing hypotheses aiming to explain the onset of autoimmune reactions are discussed in the context of genetic and environmental predisposition to type 1 diabetes (T1D). The first hypothesis has evolved along characterization of the mechanisms of self-discrimination and attributes diabetic autoimmunity to escape of reactive T cells from central regulation in the thymus. The second considers frequent occurrence of autoimmune reactions within the immune homunculus, which are adequately suppressed by regulatory T cells originating from the thymus, and occasionally, insufficient suppression results in autoimmunity. Besides thymic dysfunction, deregulation of both effector and suppressor cells can in fact result from homeostatic aberrations at the peripheral level during initial stages of evolution of adaptive immunity. Pathogenic cells sensitized in the islets are efficiently expanded in the target tissue and pancreatic lymph nodes of lymphopenic neonates. In parallel, the same mechanisms of peripheral sensitization contribute to tolerization through education of naïve/effector T cells and expansion of regulatory T cells. Experimental evidence presented for each individual mechanism implies that T1D may result from a primary effector or suppressor immune abnormality. Disturbed self-tolerance leading to T1D may well result from peripheral deregulation of innate and adaptive immunity, with variable contribution of central thymic dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoantígenos/inmunología , Autoinmunidad , Tolerancia Central , Interacción Gen-Ambiente , Humanos , Tolerancia Periférica
10.
Sleep Med Clin ; 10(3): 343-50, xv, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26329444

RESUMEN

Adopting prior models of sleep-wake transitions, a flip-flop switch in synchronized neurotransmitter activity is proposed to underlie restless leg syndrome onset. In this model, leg quiescence homeostasis sustained through concerted activities of several neurotransmitters in basal ganglia is perturbed and produces striatal motor activity along sensory activity associated with thalamocortical circuits (conscious urge and discomfort). This model explains the association of restless leg syndrome with a wide variety of associated pathologies emphasizing that perturbed function and imbalance may occur under different steady states of neurotransmitter levels. Likewise, this concept links various central and peripheral etiologies and integrates the augmenting and transient effects of therapeutic neuromodulators.


Asunto(s)
Síndrome de las Piernas Inquietas/complicaciones , Síndrome de las Piernas Inquietas/fisiopatología , Humanos , Neurotransmisores/metabolismo , Sueño/fisiología , Vigilia/fisiología
11.
Stem Cells Dev ; 24(19): 2297-306, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26067874

RESUMEN

Small-sized adult bone marrow cells isolated by counterflow centrifugal elutriation and depleted of lineage markers (Fr25lin(-)) have the capacity to differentiate into insulin-producing cells and stabilize glycemic control. This study assessed competitive migration of syngeneic stem cells to the bone marrow and islets in a murine model of chemical diabetes. VLA-4 is expressed in ∼ 25% of these cells, whereas CXCR4 is not detected, however, it is transcriptionally upregulated (6-fold). The possibility to enrich stem cells by a bone marrow homing (BM-H) functional assay was assessed in sequential transplants. Fr25lin(-) cells labeled with PKH26 were grafted into primary myeloablated recipients, and mitotically quiescent Fr25lin(-)PKH(bright) cells were sorted from the bone marrow after 2 days. The contribution of bone marrow-homed stem cells was remarkably higher in secondary recipients compared to freshly elutriated cells. The therapeutic efficacy was further increased by omission of irradiation in the secondary recipients, showing a 25-fold enrichment of islet-reconstituting cells by the bone marrow homing assay. Donor cells identified by the green fluorescent protein (GFP) and a genomic marker in sex-mismatched transplants upregulated PDX-1 and produced proinsulin, affirming the capacity of BM-H cells to convert in the injured islets. There was no evidence of transcriptional priming of freshly elutriated subsets to express PDX-1, insulin, and other markers of endocrine progenitors, indicating that the bone marrow harbors stem cells with versatile differentiation capacity. Affinity to the bone marrow can be used to enrich stem cells for pancreatic regeneration, and reciprocally, conditioning reduces the competitive incorporation in the injured islets.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Células Secretoras de Insulina/citología , Células Madre/citología , Animales , Glucemia/metabolismo , Western Blotting , Médula Ósea/metabolismo , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea/métodos , Movimiento Celular/efectos de la radiación , Células Cultivadas , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/terapia , Femenino , Expresión Génica , Proteínas de Homeodominio/genética , Hibridación Fluorescente in Situ , Insulina/sangre , Insulina/genética , Células Secretoras de Insulina/metabolismo , Integrina alfa4beta1/genética , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Transgénicos , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Transactivadores/genética
12.
Int Rev Immunol ; 34(6): 523-37, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25793647

RESUMEN

The prevalent current approach to type 1 diabetes (T1D) is the abrogation of pathogenic potential by immunosuppressive therapy, an intuitive approach aiming to slow down disease progression by the reduction of pathogenic burden. In spite of promising initial results in rodent models, there has been little efficacy of most lymphoreductive strategies in human subjects. Our analysis suggests that lymphopenia is the common denominator of ineffective immunosuppressive therapies: Immune rebound from lymphopenia is associated per se with increased susceptibility to immune reactivity, including relapse of autoimmunity. In addition, immune homeostasis and self-tolerance are not restored. These considerations raise the following question: What is the allowed degree of immunosuppressive therapy that does not elicit recurrent autoimmunity. More effective therapeutic strategies include targeted deletion of pathogenic cells, preferably in the pancreatic islets and regional lymphatics using selective T cell activation markers, re-education and remodeling of effector responses.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Terapia de Inmunosupresión , Inmunosupresores/uso terapéutico , Animales , Anticuerpos Bloqueadores/farmacología , Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Trasplante de Células , Quimioradioterapia/métodos , Estudios Clínicos como Asunto , Terapia Combinada , Modelos Animales de Enfermedad , Humanos , Inmunomodulación/efectos de los fármacos , Terapia de Inmunosupresión/métodos , Inmunosupresores/farmacología , Depleción Linfocítica , Ratones , Pronóstico , Recurrencia , Resultado del Tratamiento
13.
Blood Rev ; 29(1): 11-5, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25440916

RESUMEN

Enhanced hematopoiesis accompanies systemic responses to injury and infection. Tumor necrosis factor (TNF) produced by injured cells and interferons (IFNs) secreted by inflammatory cells is a co-product of the process of clearance of debris and removal of still viable but dysfunctional cells. Concomitantly, these cytokines induce hematopoietic stem and progenitor cell (HSPC) activity as an intrinsic component of the systemic response. The proposed scenario includes induction of HSPC activity by type I (IFNα/ß) and II (IFNγ) receptors within the quiescent bone marrow niches rendering progenitors responsive to additional signals. TNFα converges as a non-selective stimulant of HSPC activity and both cytokines synergize with other growth factors in promoting differentiation. These physiological signaling pathways of stress hematopoiesis occur quite frequent and do not cause HSPC extinction. The proposed role of IFNs and TNFs in stress hematopoiesis commends revision of their alleged involvement in bone marrow failure syndromes.


Asunto(s)
Hematopoyesis , Inflamación/metabolismo , Interferones/metabolismo , Factores de Necrosis Tumoral/metabolismo , Enfermedades Hematológicas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Transducción de Señal , Estrés Fisiológico
14.
Cell Transplant ; 24(5): 879-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24380400

RESUMEN

We have recently reported that small-sized bone marrow cells (BMCs) isolated by counterflow centrifugal elutriation and depleted of lineage markers (Fr25lin(-)) have the capacity to differentiate and contribute to regeneration of injured islets. In this study, we assess some of the characteristics of these cells compared to elutriated hematopoietic progenitors (R/O) and whole BMCs in a murine model of streptozotocin-induced chemical diabetes. The GFP(bright)CD45(+) progeny of whole BMCs and R/O progenitors progressively infiltrate the pancreas with evolution of donor chimerism; are found at islet perimeter, vascular, and ductal walls; and have a modest impact on islet recovery from injury. In contrast, Fr25lin(-) cells incorporate in the islets, convert to GFP(dim)CD45(-)PDX-1(+) phenotypes, produce proinsulin, and secrete insulin with significant contribution to stabilization of glucose homeostasis. The elutriated Fr25lin(-) cells express low levels of CD45 and are negative for SCA-1 and c-kit, as removal of cells expressing these markers did not impair conversion to produce insulin. BMCs mediate two synergistic mechanisms that contribute to islet recovery from injury: support of islet remodeling by hematopoietic cells and neogenesis of insulin-producing cells from stem cells.


Asunto(s)
Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Diferenciación Celular , Diabetes Mellitus Experimental/terapia , Células Secretoras de Insulina/metabolismo , Quimera por Trasplante/metabolismo , Animales , Diabetes Mellitus Experimental/metabolismo , Ratones , Ratones Transgénicos
15.
Front Immunol ; 5: 215, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24904571

RESUMEN

Prophylactic approaches to graft versus host disease (GvHD) have employed both phenotypic reduction of T cells and selective elimination of host-primed donor T cells in vitro and in vivo. An additional approach to GvHD prophylaxis by functional depletion of apoptosis-sensitive donor T cells without host-specific sensitization ex vivo showed remarkable reduction in GHD incidence and severity. We address the role and significance of antigen-specific sensitization of donor T cells and discuss the mechanisms of functional T cell purging by apoptosis for GvHD prevention. Host-specific sensitization is dispensable because migration is antigen-independent and donor T cell sensitization is mediated by multiple and redundant mechanisms of presentation of major and minor histocompatibility complex and tissue antigens by donor and host antigen-presenting cells. Our data suggest that potential murine and human GvH effectors reside within subsets of preactivated T cells susceptible to negative regulation by apoptosis prior to encounter of and sensitization to specific antigens.

16.
Blood ; 124(2): 176-83, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-24859365

RESUMEN

Secretion of ligands of the tumor necrosis factor (TNF) superfamily is a conserved response of parenchymal tissues to injury and inflammation that commonly perpetuates elimination of dysfunctional cellular components by apoptosis. The same signals of tissue injury that induce apoptosis in somatic cells activate stem cells and initiate the process of tissue regeneration as a coupling mechanism of injury and recovery. Hematopoietic stem and progenitor cells upregulate the TNF family receptors under stress conditions and are transduced with trophic signals. The progeny gradually acquires sensitivity to receptor-mediated apoptosis along the differentiation process, which becomes the major mechanism of negative regulation of mature proliferating hematopoietic lineages and immune homeostasis. Receptor/ligand interactions of the TNF family are physiological mechanisms transducing the need for repair, which may be harnessed in pathological conditions and transplantation. Because these interactions are physiological mechanisms of injury, neutralization of these pathways has to be carefully considered in disorders that do not involve intrinsic aberrations of excessive susceptibility to apoptosis.


Asunto(s)
Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/genética , Proliferación Celular , Células Madre Hematopoyéticas/fisiología , Humanos , Ligandos , Familia de Multigenes , Unión Proteica , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal/genética
17.
Immunology ; 142(3): 465-73, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24601987

RESUMEN

The non-obese diabetic (NOD) mouse is a prevalent disease model of type 1 diabetes. Immune aberrations that cause and propagate autoimmune insulitis in these mice are being continually debated, with evidence supporting both dominance of effector cells and insufficiency of suppressor mechanisms. In this study we assessed the behaviour of NOD lymphocytes under extreme expansion conditions using adoptive transfer into immunocompromised NOD.SCID (severe combined immunodeficiency) mice. CD4(+)  CD25(+) T cells do not cause islet inflammation, whereas splenocytes and CD4(+)  CD25(-) T cells induce pancreatic inflammation and hyperglycaemia in 80-100% of the NOD.SCID recipients. Adoptively transferred effector T cells migrate to the lymphoid organs and pancreas, proliferate, are activated in the target organ in situ and initiate inflammatory insulitis. Reconstitution of all components of the CD4(+) subset emphasizes the plastic capacity of different cell types to adopt effector and suppressor phenotypes. Furthermore, similar immune profiles of diabetic and euglycaemic NOD.SCID recipients demonstrate dissociation between fractional expression of CD25 and FoxP3 and the severity of insulitis. There were no evident and consistent differences in diabetogenic activity and immune reconstituting activity of T cells from pre-diabetic (11 weeks) and new onset diabetic NOD females. Similarities in immune phenotypes and variable distribution of effector and suppressor subsets in various stages of inflammation commend caution in interpretation of quantitative and qualitative aberrations as markers of disease severity in adoptive transfer experiments.


Asunto(s)
Traslado Adoptivo , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Huésped Inmunocomprometido , Factores de Edad , Animales , Inmunofenotipificación , Ratones , Ratones Endogámicos NOD , Ratones SCID , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
18.
Stem Cells Dev ; 23(6): 676-86, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24200242

RESUMEN

Umbilical cord blood (UCB) is a good source of hematopoietic progenitors with increasing implementation in the clinical transplant setting. This study evaluates the molecular mechanisms of progenitor resistance to apoptosis triggered by Fas cross-linking. CD34(+) and lineage-negative progenitors survive short-term ex vivo incubation and are not induced into apoptosis by Fas cross-linking. Furthermore, brief exposure of UCB cells to Fas-ligand for 24-48 h does not impair quantitative severe combine immune deficiency (SCID) reconstitution activity and appears to foster myelomonocyte reconstitution. The transcriptome of Fas receptor-positive CD34(+) cells that survived an apoptotic challenge showed significant transcriptional upregulation of caspase-8, mucosa-associated lymphoid tissue lymphoma translocation gene-1 (MALT1), HtrA2, and GSK3ß in addition to higher levels of c-FLICE inhibitory protein (FLIP), Bcl-2, and cytosolic inhibitor of apoptosis protein (cIAP) in all Fas-positive cells. Most prominent is the transcriptional upregulation of several key components the NFκB1 pathway including the membrane receptors TGF-ß, interleukin-1 (IL-1), and TCR, the associated factor TNF receptor-associated factor-6 (TRAF6), and the converting enzymes TGF-ß-activated kinase-1 (TAK1), double-stranded RNA-activated protein kinase (PKR), and α-catalytic subunit of IκB kinase (IKKα), that promote activation and nuclear translocation of this transcription factor. These data indicate that hematopoietic progenitors are not insensitive to apoptosis but are actively shielded from the extrinsic and intrinsic apoptotic pathways. This may occur through inherent transcriptional upregulation of the entire NFκB pathway in the presence of competent apoptotic signaling.


Asunto(s)
Apoptosis , Proteína Ligando Fas/metabolismo , Células Madre Hematopoyéticas/citología , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Caspasas/metabolismo , Células Cultivadas , Células Madre Hematopoyéticas/metabolismo , Humanos , FN-kappa B/efectos de los fármacos , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptor fas/metabolismo
19.
Autoimmunity ; 47(2): 105-12, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24328490

RESUMEN

Immune profiling of non-obese diabetic (NOD) is a widely employed tool to assess the mechanisms of inflammatory insulitis. Our analysis of the female NOD colony revealed similar distribution of lymphoid lineages to wild type mice, and at various ages of prediabetic and diabetic mice. The profiles of mesenteric and pancreatic lymph nodes differ and often change reciprocally due to directed migration of T cells towards the site of inflammation. Significant events in our colony include early decline in CD4(+)CD25(+)CD62L(+) Treg, accompanied by gradual increase in CD4(+)CD25(+)FoxP3(+) Treg in peripheral lymphoid organs and pancreatic infiltrates. Impressively, aged euglycemic mice display significant transient rise in CD4(+)CD25(-)FoxP3(+) Treg in the thymus, pancreas and draining lymph nodes. A significant difference was superior viability of effector and suppressor cells from new onset diabetics in the presence of high interleukin-2 (IL-2) concentrations in vitro as compared to cells of prediabetic mice. Overall, we found no correlation between FoxP3(+) Treg in the pancreatic lymph nodes and the inflammatory scores of individual NOD mice. CD25(-)FoxP3(+) Treg are markedly increased in the pancreatic infiltrates in late stages of inflammation, possibly an effort to counteract destructive insulitis. Considering extensive evidence that Treg in aged NOD mice are functionally sufficient, quantitative profiling evolves as an unreliable tool to assess mechanism and causes of inflammation under baseline conditions. Immune profiles are modulated by thymic output, cell migration, shedding of markers, proliferation, survival and in-situ evolution of regulatory cells.


Asunto(s)
Envejecimiento/inmunología , Diabetes Mellitus Tipo 1/inmunología , Hiperglucemia/inmunología , Ganglios Linfáticos/inmunología , Linfocitos T Reguladores/inmunología , Envejecimiento/sangre , Envejecimiento/patología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Glucemia/metabolismo , Movimiento Celular , Células Cultivadas , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/patología , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Expresión Génica , Humanos , Hiperglucemia/sangre , Hiperglucemia/patología , Inmunofenotipificación , Interleucina-2/farmacología , Ganglios Linfáticos/patología , Recuento de Linfocitos , Mesenterio/inmunología , Mesenterio/patología , Ratones , Ratones Endogámicos NOD , Páncreas/inmunología , Páncreas/patología , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/patología , Timo/inmunología , Timo/patología
20.
Immunol Res ; 58(1): 101-5, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24371009

RESUMEN

One of the therapeutic approaches to type 1 diabetes (T1D) focuses on enhancement of regulatory T cell (Treg) activity, either by adoptive transfer or supplementation of supporting cytokines such as interleukin-2 (IL-2). In principle, this therapeutic design would greatly benefit of concomitant reduction in pathogenic cell burden. Experimental evidence indicates that physiological recovery from lymphopenia is dominated by evolution of effector and cytotoxic cells, which abolishes the therapeutic efficacy of Treg cells. Targeted and selective depletion of effector T cells has been achieved with killer Treg using Fas ligand protein and a fusion protein composed of IL-2 and caspase-3, which showed remarkable efficacy in modulating the course of inflammatory insulitis in NOD mice. We emphasize a critical consideration in design of therapeutic approaches to T1D, immunomodulation without lymphoreduction to avoid the detrimental consequences of rebound recovery from lymphopenia.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Linfopenia/inmunología , Linfocitos T Reguladores/inmunología , Animales , Caspasa 3/inmunología , Diabetes Mellitus Tipo 1/terapia , Humanos , Inmunomodulación , Interleucina-2/inmunología , Ratones Endogámicos NOD
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...