Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 140(12): 1431-1447, 2022 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-35443019

RESUMEN

Vasoactive intestinal polypeptide (VIP), an anti-inflammatory neuropeptide with pleiotropic cardiovascular effects, induces differentiation of hematopoietic stem cells into regulatory dendritic cells that limit graft-versus-host disease (GVHD) in allogeneic hematopoietic stem cell transplant (HSCT) recipients. We have previously shown that donor plasmacytoid dendritic cells (pDCs) in bone marrow (BM) donor grafts limit the pathogenesis of GVHD. In this current study we show that murine and human pDCs express VIP, and that VIP-expressing pDCs limit T-cell activation and expansion using both in vivo and in vitro model systems. Using T cells or pDCs from transgenic luciferase+ donors in murine bone marrow transplantation (BMT), we show similar homing patterns of donor pDCs and T cells to the major sites for alloactivation of donor T cells: spleen and gut. Cotransplanting VIP-knockout (KO) pDCs with hematopoietic stem cells and T cells in major histocompatibility complex mismatched allogeneic BMT led to lower survival, higher GVHD scores, and more colon crypt cell apoptosis than transplanting wild-type pDCs. BMT recipients of VIP-KO pDCs had more T helper 1 polarized T cells, and higher plasma levels of granulocyte-macrophage colony-stimulating factor and tumor necrosis factor-α than recipients of wild-type pDCs. T cells from VIP-KO pDC recipients had increasing levels of bhlhe40 transcripts during the first 2 weeks posttransplant, and higher levels of CyclophilinA/Ppia transcripts at day 15 compared with T cells from recipients of wild-type pDCs. Collectively, these data indicate paracrine VIP synthesis by donor pDCs limits pathogenic T-cell inflammation, supporting a novel mechanism by which donor immune cells regulate T-cell activation and GVHD in allogeneic BMT.


Asunto(s)
Enfermedad Injerto contra Huésped , Animales , Trasplante de Médula Ósea/efectos adversos , Células Dendríticas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33649184

RESUMEN

Kv1.3 potassium channels, expressed by proinflammatory central nervous system mononuclear phagocytes (CNS-MPs), are promising therapeutic targets for modulating neuroinflammation in Alzheimer's disease (AD). The molecular characteristics of Kv1.3-high CNS-MPs and their cellular origin from microglia or CNS-infiltrating monocytes are unclear. While Kv1.3 blockade reduces amyloid beta (Aß) burden in mouse models, the downstream immune effects on molecular profiles of CNS-MPs remain unknown. We show that functional Kv1.3 channels are selectively expressed by a subset of CD11b+CD45+ CNS-MPs acutely isolated from an Aß mouse model (5xFAD) as well as fresh postmortem human AD brain. Transcriptomic profiling of purified CD11b+Kv1.3+ CNS-MPs, CD11b+CD45int Kv1.3neg microglia, and peripheral monocytes from 5xFAD mice revealed that Kv1.3-high CNS-MPs highly express canonical microglial markers (Tmem119, P2ry12) and are distinct from peripheral Ly6chigh/Ly6clow monocytes. Unlike homeostatic microglia, Kv1.3-high CNS-MPs express relatively lower levels of homeostatic genes, higher levels of CD11c, and increased levels of glutamatergic transcripts, potentially representing phagocytic uptake of neuronal elements. Using irradiation bone marrow CD45.1/CD45.2 chimerism in 5xFAD mice, we show that Kv1.3+ CNS-MPs originate from microglia and not blood-derived monocytes. We show that Kv1.3 channels regulate membrane potential and early signaling events in microglia. Finally, in vivo blockade of Kv1.3 channels in 5xFAD mice by ShK-223 reduced Aß burden, increased CD11c+ CNS-MPs, and expression of phagocytic genes while suppressing proinflammatory genes (IL1b). Our results confirm the microglial origin and identify unique molecular features of Kv1.3-expressing CNS-MPs. In addition, we provide evidence for CNS immunomodulation by Kv1.3 blockers in AD mouse models resulting in a prophagocytic phenotype.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Canal de Potasio Kv1.3/metabolismo , Microglía/metabolismo , Células Mieloides/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Canal de Potasio Kv1.3/genética , Masculino , Ratones
3.
Blood ; 132(23): 2506-2519, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30257880

RESUMEN

The intestinal microbiota in allogeneic bone marrow transplant (allo-BMT) recipients modulates graft-versus-host disease (GVHD), a systemic inflammatory state initiated by donor T cells that leads to colitis, a key determinant of GVHD severity. Indole or indole derivatives produced by tryptophan metabolism in the intestinal microbiota limit intestinal inflammation caused by diverse stressors, so we tested their capacity to protect against GVHD in murine major histocompatibility complex-mismatched models of allo-BMT. Indole effects were assessed by colonization of allo-BMT recipient mice with tryptophanase positive or negative strains of Escherichia coli, or, alternatively, by exogenous administration of indole-3-carboxaldehyde (ICA), an indole derivative. Treatment with ICA limited gut epithelial damage, reduced transepithelial bacterial translocation, and decreased inflammatory cytokine production, reducing GVHD pathology and GVHD mortality, but did not compromise donor T-cell-mediated graft-versus-leukemia responses. ICA treatment also led to recipient-strain-specific tolerance of engrafted T cells. Transcriptional profiling and gene ontology analysis indicated that ICA administration upregulated genes associated with the type I interferon (IFN1) response, which has been shown to protect against radiation-induced intestinal damage and reduce subsequent GVHD pathology. Accordingly, protective effects of ICA following radiation exposure were abrogated in mice lacking IFN1 signaling. Taken together, these data indicate that indole metabolites produced by the intestinal microbiota act via type I IFNs to limit intestinal inflammation and damage associated with myeloablative chemotherapy or radiation exposure and acute GVHD, but preserve antitumor responses, and may provide a therapeutic option for BMT patients at risk for GVHD.


Asunto(s)
Trasplante de Médula Ósea , Escherichia coli/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedad Injerto contra Huésped , Indoles , Interferón Tipo I/metabolismo , Mucosa Intestinal , Aloinjertos , Animales , Traslocación Bacteriana/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/microbiología , Indoles/farmacocinética , Indoles/farmacología , Interferón Tipo I/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados
4.
Biol Blood Marrow Transplant ; 24(12): 2373-2380, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29981848

RESUMEN

Extracorporeal photopheresis (ECP) is an accepted treatment for chronic graft-versus-host disease (cGVHD); however, the mechanism of action is unclear. We conducted a prospective multicenter clinical trial to assess ECP response rates using the 2005 National Institutes of Health (NIH) consensus criteria and to assess the relationship between regulatory T cells (Tregs) and treatment response (NCT01324908). Eighty-three patients with any NIH subtype of cGVHD were enrolled, irrespective of number of prior lines of treatment, and 6 were subsequently excluded because of the absence of follow-up from cancer relapse, infection, or study withdrawal. Study outcomes were provider-assessed response and formal response by 2005 NIH criteria. Peripheral blood samples were collected at prespecified study visits and were analyzed by flow cytometry for Tregs. In a heavily pretreated cohort of patients, with a median of 2 prior lines of therapy, 62.3% of patients had a provider-assessed response to ECP and 43.5% had response by NIH criteria. These assessments showed only a slight agreement (kappa statistic, .09). In a logistic regression model that included previously identified risk factors such as bilirubin, platelet count, and time from transplant to study entry, no clinical factors were associated with the provider's response assessment. Furthermore, there was no significant difference in percentage of Tregs in blood leukocytes at study entry and completion or in overall change in Treg frequency between ECP responders and nonresponders. ECP was associated with a clinically significant decrease in median prednisone dose (.36 to .14 mg/kg, P < .001) from study entry to last visit and a significant decrease in global severity of cGVHD and total body surface area with erythematous rash. Overall, ECP was able to deliver response using NIH response criteria in a highly pretreated cohort with moderate and severe cGVHD independent of most previous risk factors for adverse outcomes of cGVHD.


Asunto(s)
Enfermedad Injerto contra Huésped/terapia , Fotoféresis/métodos , Linfocitos T Reguladores/metabolismo , Anciano , Enfermedad Crónica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos
5.
Biol Blood Marrow Transplant ; 24(5): 973-982, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29307717

RESUMEN

More than 90% of allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients receive red blood cell (RBC) or platelet transfusions in the peritransplantation period. We tested the hypothesis that transfusions are associated with the development of severe (grade III-IV) acute graft-versus-host disease (aGVHD) or mortality after allo-HSCT in a retrospective study of 322 consecutive patients receiving an allogeneic bone marrow or granulocyte colony-stimulating factor-mobilized blood stem cell graft for a hematologic malignancy. Counting transfused RBC and platelet units between day -7 pretransplantation and day +27 post-transplantation, but excluding transfusions administered after a diagnosis of aGVHD, yielded medians of 5 RBC units and 2 platelet units transfused. Sixty-three patients (20%) developed a maximal grade III-IV aGVHD with onset up to day +150 post-transplantation (median aGVHD onset of 28 days). HLA mismatch (hazard ratio [HR], 2.4; 95% confidence interval [CI], 1.2 to 4.7; P = .01), and transfusion of more than the median number of RBC units (HR, 2.1; 95% CI, 1.1 to 3.7; P = .02) were independently associated with greater risk of grade III-IV aGVHD in a multivariable analysis model. Disease risk strata (HR, 1.7; 95% CI, 1.2 to 2.4 for high risk versus low risk; P = .005) and transfusion of more than the median number of RBC units (HR, 1.4; 95% CI, 1.0 to 2.0; P = .054) were independently associated with inferior overall survival. These data support our hypothesis that peritransplantation RBC transfusions are associated with the risk of developing severe aGVHD and worse overall survival following allo-HSCT, and suggest that strategies to reduce routine RBC transfusion may favorably reduce the incidence and severity of GVHD.


Asunto(s)
Transfusión de Eritrocitos/efectos adversos , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Adulto , Anciano , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Masculino , Persona de Mediana Edad , Atención Perioperativa/efectos adversos , Atención Perioperativa/métodos , Transfusión de Plaquetas , Estudios Retrospectivos , Riesgo , Análisis de Supervivencia , Trasplante Homólogo/efectos adversos , Adulto Joven
6.
Blood ; 128(21): 2568-2579, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27758873

RESUMEN

One of the central challenges of transplantation is the development of alloreactivity despite the use of multiagent immunoprophylaxis. Effective control of this immune suppression-resistant T-cell activation represents one of the key unmet needs in the fields of both solid-organ and hematopoietic stem cell transplant (HCT). To address this unmet need, we have used a highly translational nonhuman primate (NHP) model to interrogate the transcriptional signature of T cells during breakthrough acute graft-versus-host disease (GVHD) that occurs in the setting of clinically relevant immune suppression and compared this to the hyperacute GVHD, which develops in unprophylaxed or suboptimally prophylaxed transplant recipients. Our results demonstrate the complex character of the alloreactivity that develops during ongoing immunoprophylaxis and identify 3 key transcriptional hallmarks of breakthrough acute GVHD that are not observed in hyperacute GVHD: (1) T-cell persistence rather than proliferation, (2) evidence for highly inflammatory transcriptional programming, and (3) skewing toward a T helper (Th)/T cytotoxic (Tc)17 transcriptional program. Importantly, the gene coexpression profiles from human HCT recipients who developed GVHD while on immunosuppressive prophylactic agents recapitulated the patterns observed in NHP, and demonstrated an evolution toward a more inflammatory signature as time posttransplant progressed. These results strongly implicate the evolution of both inflammatory and interleukin 17-based immune pathogenesis in GVHD, and provide the first map of this evolving process in primates in the setting of clinically relevant immunomodulation. This map represents a novel transcriptomic resource for further systems-based efforts to study the breakthrough alloresponse that occurs posttransplant despite immunoprophylaxis and to develop evidence-based strategies for effective treatment of this disease.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Interleucina-17/inmunología , Linfocitos T Citotóxicos , Linfocitos T Colaboradores-Inductores , Enfermedad Aguda , Aloinjertos , Animales , Modelos Animales de Enfermedad , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/prevención & control , Haplorrinos , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/terapia , Masculino , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología
7.
PLoS Pathog ; 11(3): e1004770, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25822986

RESUMEN

Imatinib mesylate (Gleevec) inhibits Abl1, c-Kit, and related protein tyrosine kinases (PTKs) and serves as a therapeutic for chronic myelogenous leukemia and gastrointestinal stromal tumors. Imatinib also has efficacy against various pathogens, including pathogenic mycobacteria, where it decreases bacterial load in mice, albeit at doses below those used for treating cancer. We report that imatinib at such low doses unexpectedly induces differentiation of hematopoietic stem cells and progenitors in the bone marrow, augments myelopoiesis but not lymphopoiesis, and increases numbers of myeloid cells in blood and spleen. Whereas progenitor differentiation relies on partial inhibition of c-Kit by imatinib, lineage commitment depends upon inhibition of other PTKs. Thus, imatinib mimics "emergency hematopoiesis," a physiological innate immune response to infection. Increasing neutrophil numbers by adoptive transfer sufficed to reduce mycobacterial load, and imatinib reduced bacterial load of Franciscella spp., which do not utilize imatinib-sensitive PTKs for pathogenesis. Thus, potentiation of the immune response by imatinib at low doses may facilitate clearance of diverse microbial pathogens.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Francisella/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Mesilato de Imatinib/farmacología , Mielopoyesis/efectos de los fármacos , Neutrófilos/inmunología , Animales , Diferenciación Celular/inmunología , Recuento de Leucocitos , Ratones , Mielopoyesis/inmunología
9.
Blood ; 119(4): 1075-85, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22130799

RESUMEN

Allogeneic hematopoietic stem cell transplantation (HSCT) can eradicate chemorefractory leukemia through the graft-versus-leukemia (GVL) activity of donor T cells. However, the clinical success of allo-HSCT is limited by the graft-versus-host disease (GVHD) activity of donor T cells. We have reported previously that donor bone marrow precursors of plasmacytoid dendritic cells (pre-pDCs) can activate donor T cells toward T-helper 1 immune polarization in murine allogeneic HSCT. To optimize the GVL activity of these activated donor T cells and limit their graft versus host activity, we engineered the cellular constituents of an allogeneic hematopoietic stem cell graft with highly purified hematopoietic stem cells, T cells, and pre-pDCs and studied their GVL and GVHD activities in a murine model of allogeneic HSCT. Transplanted donor pre-pDCs expanded in vivo for 2 weeks after transplant, and they markedly augmented the activation and GVL activity of donor T cells while attenuating their GVHD activity, leading to an improved therapeutic index. Bidirectional signaling between donor T cells and donor pDCs with IFN-γ synthesis by donor T cells inducing indoleamine 2,3-dioxygenase synthesis by donor pDCs limited GVHD by altering the balance between donor T-reg and inflammatory T cells. Manipulating the content of donor DC precursors in allogeneic HSCT is a novel method to optimize the balance between GVL and GVHD.


Asunto(s)
Comunicación Celular , Células Dendríticas/metabolismo , Enfermedad Injerto contra Huésped/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo , Leucemia de Células T/inmunología , Linfocitos T/metabolismo , Animales , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Regulación Enzimológica de la Expresión Génica , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Interferón gamma/genética , Leucemia de Células T/metabolismo , Leucemia de Células T/terapia , Activación de Linfocitos , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Inmunológicos , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Trasplante Homólogo , Trasplante Isogénico , Receptor de Interferón gamma
10.
J Immunol ; 187(2): 1057-65, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21677142

RESUMEN

Vasoactive intestinal peptide (VIP) induces regulatory dendritic cells (DC) in vitro that inhibit cellular immune responses. We tested the role of physiological levels of VIP on immune responses to murine CMV (mCMV) using VIP-knockout (VIP-KO) mice and radiation chimeras engrafted with syngenic VIP-KO hematopoietic cells. VIP-KO mice had less weight loss and better survival following mCMV infection compared with wild-type (WT) littermates. mCMV-infected VIP-KO mice had lower viral loads, faster clearance of virus, with increased numbers of IFN-γ(+) NK and NKT cells, and enhanced cytolytic activity of NK cells. Adaptive antiviral cellular immunity was increased in mCMV-infected VIP-KO mice compared with WT mice, with more Th1/Tc1-polarized T cells, fewer IL-10(+) T cells, and more mCMV-M45 epitope peptide MHC class I tetramer(+) CD8(+) T cells (tetramer(+) CD8 T cells). mCMV-immune VIP-KO mice had enhanced ability to clear mCMV peptide-pulsed target cells in vivo. Enhanced antiviral immunity was also seen in WT transplant recipients engrafted with VIP-KO hematopoietic cells, indicating that VIP synthesized by neuronal cells did not suppress immune responses. Following mCMV infection there was a marked upregulation of MHC-II and CD80 costimulatory molecule expression on DC from VIP-KO mice compared with DC from WT mice, whereas programmed death-1 and programmed death ligand-1 expression were upregulated in activated CD8(+) T cells and DC, respectively, in WT mice, but not in VIP-KO mice. Because the absence of VIP in immune cells increased innate and adaptive antiviral immunity by altering costimulatory and coinhibitory pathways, selective targeting of VIP signaling represents an attractive therapeutic target to enhance antiviral immunity.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Polaridad Celular/inmunología , Células Madre Hematopoyéticas/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/prevención & control , Muromegalovirus/inmunología , Células TH1/inmunología , Péptido Intestinal Vasoactivo/deficiencia , Animales , Trasplante de Médula Ósea/patología , Polaridad Celular/genética , Femenino , Células Madre Hematopoyéticas/patología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inmunización Secundaria , Inmunofenotipificación , Listeriosis/inmunología , Listeriosis/patología , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Quimera por Radiación , Células TH1/microbiología , Células TH1/virología , Péptido Intestinal Vasoactivo/biosíntesis , Péptido Intestinal Vasoactivo/genética
11.
Front Immunol ; 13: 959658, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36189324

RESUMEN

Despite the curative potential of hematopoietic cell transplantation (HCT) for hematologic malignancies, graft-versus-host disease (GVHD) remains a substantial cause of morbidity and mortality, particularly if treatment is refractory. Treatment with additional immunosuppression including steroids often leads to opportunistic infections and organ dysfunction. Novel therapies are greatly needed, specifically ones that lead to responses in treatment-refractory patients and are better tolerated. Mesenchymal stromal cells (MSCs) are non-hematopoietic tolerogenic cells present in normal bone marrow (BM), which can be expanded ex vivo to therapeutic doses. Their safety and efficacy have been assessed in inflammatory disorders including GVHD, but heterogeneity in clinical responses has led some to examine MSC manufacturing and administration procedures, which may impact in vivo efficacy. We hypothesized that autologous, early-passage, and culture-recovered (after freeze and thaw) MSCs would be safe and may have superior efficacy. In this phase I single-center trial, we assessed MSC safety and early efficacy of an escalating number of doses (2 × 106/kg doses; dose level 1, single dose; dose level 2, two weekly doses; dose level 3, four weekly doses) in patients aged ≥12 years with treatment-refractory acute or chronic GVHD. Eleven enrolled patients received some or all planned MSC infusions, with a median age at enrollment of 37 years. The most common primary HCT indication was leukemia, and the median time from HCT to first MSC infusion was 2.6 years. MSC infusion was well tolerated, with all severe adverse events expected and determined to be unlikely or definitely not related to the study. Thus, no dose-limiting toxicities occurred in the three dose levels. Three of four patients with acute GVHD (or overlap with acute features) had responses seen at any timepoint, ranging from partial to complete. In those with a chronic GVHD indication (n = 7), an overall response at 3 months was partial in five, stable in one, and progressive in one. No appreciable differences were seen between dose levels in peripheral blood lymphocyte subsets. In conclusion, autologous and culture-recovered MSCs were safe in the setting of refractory GVHD following HCT for hematologic malignancy, and clinical responses were most notable in patients with acute GVHD.


Asunto(s)
Enfermedad Injerto contra Huésped , Neoplasias Hematológicas , Trasplante de Células Madre Hematopoyéticas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Adulto , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/terapia , Neoplasias Hematológicas/patología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Trasplante de Células Madre Mesenquimatosas/métodos , Esteroides
12.
J Immunol ; 183(12): 7799-809, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19933853

RESUMEN

We investigated the roles of specific subsets of donor APCs purified from bone marrow in donor T cell activation and graft-vs-leukemia (GvL) activity in murine models of hemopoietic stem cell transplantation. Lineage(-)CD11c(+) APC precursors were separated from donor bone marrow based on expression of CD11b. Transplanting lineage(-)CD11c(+)CD11b(-) APC (CD11b(-) APC) in combination with c-kit(+)Sca-1(+)lineage(-) hemopoietic stem cells (HSC) and congenic donor T cells led to increased donor CD4(+) and CD8(+) T cell proliferation and higher donor T cell chimerism than with transplanting grafts containing HSC, T cells, and lineage(-)CD11c(+)CD11b(+) APCs (CD11b(+) APC), or grafts containing only HSC and T cells. Transplanting CD11b(-) APCs induced Th1/type 1 cytotoxic T lymphocyte donor T cell immune polarization and enhanced GvL activity of donor T cells without increased graft-vs-host disease in both MHC- and minor histocompatibility Ag-mismatched murine hemopoietic stem cell transplantation models, whereas CD11b(+) APCs led to Th2/type 2 cytotoxic T lymphocyte donor T cell immune polarization. Donor CD11b(-) APCs were plasmacytoid dendritic cell progenitors (>90% CD317; PDCA-1(+)) and up-regulated CD80, CD86, and IL-12 during alloantigen presentation, whereas CD11b(+) APCs expressed Gr-1 and up-regulated expression of programmed death ligands-1 and 2 after activation. These results are the first to show that manipulation of the content of donor APCs in allogeneic HSC grafts can regulate donor T cell immunity and enhance GvL without increasing graft-vs-host disease activity.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células de la Médula Ósea/inmunología , Polaridad Celular/inmunología , Efecto Injerto vs Leucemia/inmunología , Trasplante de Células Madre Hematopoyéticas , Activación de Linfocitos/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/trasplante , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Refuerzo Inmunológico de Injertos/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Isoantígenos/administración & dosificación , Isoantígenos/biosíntesis , Isoantígenos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/trasplante
13.
Transfusion ; 50(11): 2424-31, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20529004

RESUMEN

BACKGROUND: One proposed mechanism of extracorporeal photopheresis (ECP) in reducing chronic graft-versus-host disease (cGVHD) is alteration in numbers of circulating dendritic cells (DCs). This hypothesis was tested by correlating numbers of DC precursors and T cells in the blood before and during ECP therapy with response of cGVHD. STUDY DESIGN AND METHODS: Twenty-five patients with cGVHD were treated with ECP. Data were collected with emphasis on blood cellular markers, clinical response to ECP, and overall survival. RESULTS: Fourteen patients (56%) responded and had better 2-year survival than nonresponders (88% vs. 18%, p=0.003). Responders had higher baseline circulating myeloid DC (mDC) and plasmacytoid DC precursors and CD4+ and CD8+ T cells compared with nonresponders. Receiver operating characteristic curve analyses showed that the best baseline cutoff values to predict response to ECP were mDC counts of 3.7 cells/µL (79% sensitivity, 82% specificity) and CD4+ T-cell counts of 104 cells/µL (71% sensitivity, 82% specificity). CD4+ T cells declined in responders over time, but not in nonresponders, and no significant changes were seen in CD8 T-cell or DC numbers over a 12-month period in responder or nonresponder groups. CONCLUSIONS: Higher baseline numbers of circulating DCs and T cells may predict clinical response to ECP in patients with cGVHD.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Células Dendríticas/citología , Enfermedad Injerto contra Huésped/terapia , Células Madre Hematopoyéticas/citología , Fotoféresis/métodos , Adulto , Anciano , Enfermedad Crónica , Femenino , Estudios de Seguimiento , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Fotoféresis/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Valor Predictivo de las Pruebas , Curva ROC , Recurrencia , Sensibilidad y Especificidad , Adulto Joven
14.
Blood Adv ; 3(15): 2250-2263, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31345792

RESUMEN

The clinical utility of monitoring immune reconstitution after allotransplant was evaluated using data from Blood and Marrow Transplant Clinical Trials Network BMT CTN 0201 (NCT00075816), a multicenter randomized study of unrelated donor bone marrow (BM) vs granulocyte colony-stimulating factor (G-CSF)-mobilized blood stem cell (G-PB) grafts. Among 410 patients with posttransplant flow cytometry measurements of immune cell subsets, recipients of G-PB grafts had faster T-cell reconstitution than BM recipients, including more naive CD4+ T cells and T-cell receptor excision circle-positive CD4+ and CD8+ T cells at 3 months, consistent with better thymic function. Faster reconstitution of CD4+ T cells and naive CD4+ T cells at 1 month and CD8+ T cells at 3 months predicted more chronic graft-versus-host disease (GVHD) but better survival in G-PB recipients, but consistent associations of T-cell amounts with GVHD or survival were not seen in BM recipients. In contrast, a higher number of classical dendritic cells (cDCs) in blood samples at 3 months predicted better survival in BM recipients. Functional T-cell immunity measured in vitro by cytokine secretion in response to stimulation with cytomegalovirus peptides was similar when comparing blood samples from BM and G-PB recipients, but the degree to which acute GVHD suppressed immune reconstitution varied according to graft source. BM, but not G-PB, recipients with a history of grades 2-4 acute GVHD had lower numbers of B cells, plasmacytoid dendritic cells, and cDCs at 3 months. Thus, early measurements of T-cell reconstitution are predictive cellular biomarkers for long-term survival and response to GVHD therapy in G-PB recipients, whereas more robust DC reconstitution predicted better survival in BM recipients.


Asunto(s)
Trasplante de Médula Ósea , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Reconstitución Inmune , Trasplante de Médula Ósea/métodos , Trasplante de Médula Ósea/mortalidad , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/etiología , Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Cinética , Recuento de Linfocitos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Evaluación del Resultado de la Atención al Paciente , Pronóstico , Análisis de Supervivencia , Factores de Tiempo
15.
Exp Hematol ; 34(7): 895-904, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16797417

RESUMEN

OBJECTIVE: To compare the abilities of different ex vivo methods of treating donor lymphocytes to inhibit graft-vs-host disease (GVHD) while preserving graft-vs-leukemia (GVL) activity in murine models of allogeneic bone marrow transplantation. METHODS: Donor/recipient pairs included MHC fully mismatched, MHC haplomismatched, and MiHA mismatched strain combinations. T cell-depleted BM (TCD-BM) was transplanted in combination with untreated, fludarabine-treated, 7.5-Gy gamma-irradiated, or psoralen/UVA (PUVA)-treated splenocytes. GVL activity was studied by adding a lethal number of recipient-type lymphoma cells. Posttransplant survival was determined, and GVHD and GVL activity were assessed by clinical and pathological scoring. Hematopoietic chimerism and donor T-cell expansion were analyzed by flow cytometry of peripheral blood samples at 30 and 60 days posttransplant. RESULTS: Allogeneic splenocytes treated with fludarabine, 7.5 Gy gamma-irradiation, or PUVA had significantly diminished GVHD activity, and all treated donor T cells facilitated engraftment by low-dose TCD-BM. Allogeneic splenocytes treated with fludarabine and, to a lesser extent, PUVA retained GVL activity and contributed more to donor T-cell chimerism compared to gamma-irradiated donor splenocytes. CONCLUSION: Among ex vivo methods of treating donor T cells to limit their proliferative capacity, fludarabine exposure had the greatest differential ability to inhibit the GVHD activity of allogeneic lymphocytes, while preserving their GVL activity and ability to engraft recipients. Thus, ex vivo treatment with fludarabine was superior to gamma-irradiation or PUVA in separating GVL from GVHD activity in murine models of allogeneic bone marrow transplantation.


Asunto(s)
Trasplante de Médula Ósea , Efecto Injerto vs Leucemia , Vidarabina/análogos & derivados , Animales , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología , Acondicionamiento Pretrasplante , Trasplante Homólogo , Vidarabina/farmacología , Vidarabina/uso terapéutico
16.
Cancer Res ; 76(23): 6802-6815, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27671676

RESUMEN

The goal of allogeneic bone marrow transplantation (allo-BMT) is elimination of leukemia cells through the graft-versus-leukemia (GvL) activity of donor cells, while limiting graft-versus-host disease (GvHD). Immune checkpoint pathways regulate GvL and GvHD activities, but blocking antibodies or genetic inactivation of these pathways can cause lethal GVHD. Vasoactive intestinal peptide (VIP) is an immunosuppressive neuropeptide that regulates coinhibitory pathways; its role in allo-BMT has not been studied. We found VIP transiently expressed in donor NK, NK-T, dendritic cells, and T cells after allo transplant, as well as host leukocytes. A peptide antagonist of VIP signaling (VIPhyb) increased T-cell proliferation in vitro and reduced IL10 expression in donor T cells. Treatment of allo-BMT recipients with VIPhyb, or transplanting donor grafts lacking VIP (VIP-KO), activated donor T-cells in lymphoid organs, reduced T-cell homing to GvHD target organs, and enhanced GvL without increasing GvHD in multiple allo-BMT models. Genetic or ex vivo depletion of donor NK cells or CD8+ T cells from allografts abrogated the VIPhyb-enhanced GvL activity. VIPhyb treatment led to downregulation of PD-1 and PD-L1 expression on donor immune cells, increased effector molecule expression, and expanded oligoclonal CD8+ T cells that protected secondary allo transplant recipients from leukemia. Blocking VIP signaling thus represents a novel pharmacologic approach to separate GvL from GvHD and enhance adaptive T-cell responses to leukemia-associated antigens in allo-BMT. Cancer Res; 76(23); 6802-15. ©2016 AACR.


Asunto(s)
Trasplante de Médula Ósea/métodos , Efecto Injerto vs Leucemia/inmunología , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo/métodos , Péptido Intestinal Vasoactivo/metabolismo , Animales , Humanos , Masculino , Ratones , Transducción de Señal
17.
Immunol Res ; 29(1-3): 269-82, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15181288

RESUMEN

A major goal of our research is to reduce the graft-vs-host disease (GVHD) activity of allogeneic donor T cells in bone marrow transplantation (BMT), while preserving graft-vs-tumor (GVT) effects. Using ex vivo immunosuppressive strategies and cell-separation techniques to modulate the graft prior to transplantation, we examined the roles of different donor immune cells on GVHD and GVT effects in allogeneic mouse models. Our results demonstrate that donor-memory CD4 T cells facilitate posttransplant immunological reconstitution without causing GVHD, whereas transplantation of equal numbers of donor naïve CD4 T cells leads to fatal GVHD. The initial events of donor T cells interacting with antigen-presenting cells (APCs) in the transplant recipient appear to be critical to the development of GVT, GVHD, or anergy to alloantigens. In the setting of clinical BMT, increased numbers of donor type 2 dendritic cells (DCs) were associated with an increased rate of posttransplant relapse, and decreased rates of chronic GVHD. In a mouse transplant model, manipulation of the DC content of bone marrow grafts was achieved by depletion of CD11b+ cells. Mice transplanted with CD11b- depleted marrow showed enhanced posttransplant expansion of memory T cells with markedly improved GVT activity and limited GVHD compared to recipients of unmanipulated marrow. A model that differentiates GVT from GVHD based on interaction of T-cell subsets with DC subsets is proposed.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Efecto Injerto vs Tumor/inmunología , Vidarabina/análogos & derivados , Animales , Células Dendríticas/inmunología , Rayos gamma , Supervivencia de Injerto/inmunología , Enfermedad Injerto contra Huésped/inmunología , Efecto Injerto vs Tumor/efectos de los fármacos , Efecto Injerto vs Tumor/efectos de la radiación , Receptores de Hialuranos/inmunología , Ratones , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/efectos de la radiación , Trasplante Homólogo , Vidarabina/farmacología
18.
J Hematol Oncol ; 7: 16, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24580829

RESUMEN

BACKGROUND: Donor cell engraftment is critical for the success of allogeneic bone marrow transplants. Graft failure is a result of donor cells either failing to engraft initially or being eliminated at later time points. Donor cell engraftment is facilitated by donor T cells, which eliminate residual host hemato-lymphoid effector cells such as NK cells and T cells. METHODS: We aimed to explore the role of host hematopoietic cell derived IL-12 on donor cell engraftment in a murine model of BMT. We established radiation chimeras by transplanting C57BL6/J (B6) mice with BM from either congenic B6 mice or IL-12p40 KO mice. These WT → WT or IL-12 KO → WT chimeras then underwent a secondary transplant with allogeneic (FVB) BM. Survival, engraftment, donor T cell expansion, cytokine production by donor T cells, as well as expression of stimulatory markers on donor T cells was analyzed. RESULTS: Mice whose residual host hematopoietic cells were capable of producing IL-12 had modestly higher survival, higher donor T cell engraftment, and significantly higher donor erythroid engraftment. We have also found that an increased number of donor T cells in IL-12 KO → WT chimeras have a regulatory phenotype, expressing FoxP3, producing lower levels of TNF-α, higher levels of IL-10, and expressing higher levels of ICOS as well as PD-1 on CD4+ T cells. CONCLUSIONS: To our knowledge, this is the first report of a beneficial role of IL-12 production by host cells in the context of bone marrow engraftment in a murine model of BMT. These findings support the clinical use of exogenous IL-12 for use in settings where graft failure is of concern.


Asunto(s)
Trasplante de Médula Ósea/métodos , Interleucina-12/inmunología , Linfocitos T/trasplante , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Donantes de Tejidos
19.
PLoS One ; 8(7): e64837, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23843936

RESUMEN

Myeloid derived suppressor cells (MDSCs) from tumor-bearing mice are important negative regulators of anti-cancer immune responses, but the role for immature myeloid cells (IMCs) in non-tumor-bearing mice in the regulation of immune responses are poorly described. We studied the immune-suppressive activity of IMCs from the bone marrow (BM) of C57Bl/6 mice and the mechanism(s) by which they inhibit T-cell activation and proliferation. IMCs, isolated from BM by high-speed FACS, inhibited mitogen-induced proliferation of CD4(+) and CD8(+) T-cells in vitro. Cell-to-cell contact of T-cells with viable IMCs was required for suppression. Neither neutralizing antibodies to TGFß1, nor genetic disruption of indolamine 2,3-dioxygenase, abrogated IMC-mediated suppressive activity. In contrast, suppression of T-cell proliferation was absent in cultures containing IMCs from interferon-γ (IFN-γ) receptor KO mice or T-cells from IFN-γ KO mice (on the C57Bl/6 background). The addition of NO inhibitors to co-cultures of T-cells and IMC significantly reduced the suppressive activity of IMCs. IFN-γ signaling between T-cells and IMCs induced paracrine Nitric Oxide (NO) release in culture, and the degree of inhibition of T-cell proliferation was proportional to NO levels. The suppressive activity of IMCs from the bone marrow of tumor-free mice was comparable with MDSCs from BALB/c bearing mice 4T1 mammary tumors. These results indicate that IMCs have a role in regulating T-cell activation and proliferation in the BM microenvironment.


Asunto(s)
Células de la Médula Ósea/citología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Células Mieloides/citología , Óxido Nítrico/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Inhibidores Enzimáticos/farmacología , Femenino , Expresión Génica , Indolamina-Pirrol 2,3,-Dioxigenasa/deficiencia , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Interferón gamma/deficiencia , Interferón gamma/genética , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/metabolismo , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/inmunología , Transducción de Señal , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/biosíntesis
20.
Immunotherapy ; 1(4): 599-621, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20191089

RESUMEN

Routine methods to maximize the graft-versus-leukemia (GvL) activity of allogeneic hematopoietic stem cell transplantation (HSCT) without the detrimental effects of graft-versus-host disease (GvHD) are lacking. Depletion or inhibition of alloreactive T cells is partially effective in preventing GvHD, but usually leads to decreased GvL activity. The current model for the pathophysiology of acute GvHD describes a series of immune pathways that lead to activation of donor T cells and inflammatory cytokines responsible for tissue damage in acute GvHD. This model does not account for how allotransplant can lead to GvL effects without GvHD, or how the initial activation of donor immune cells may lead to counter-regulatory effects that limit GvHD. In this review, we will summarize new findings that support a more complex model for the initiation of GvHD and GvL activities in allogeneic HSCT, and discuss the potential of novel strategies to enhance GvL activity of the transplant.


Asunto(s)
Células Dendríticas/inmunología , Enfermedad Injerto contra Huésped/inmunología , Efecto Injerto vs Leucemia/inmunología , Trasplante de Células Madre Hematopoyéticas , Linfocitos T Reguladores/inmunología , Animales , Comunicación Celular , Células Dendríticas/efectos de los fármacos , Enfermedad Injerto contra Huésped/fisiopatología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Activación de Linfocitos/efectos de los fármacos , Depleción Linfocítica , Linfocitos T Reguladores/efectos de los fármacos , Balance Th1 - Th2 , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA