Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Blood ; 129(26): 3428-3439, 2017 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-28533309

RESUMEN

The aryl hydrocarbon receptor (AHR) plays an important physiological role in hematopoiesis. AHR is highly expressed in hematopoietic stem and progenitor cells (HSPCs) and inhibition of AHR results in a marked expansion of human umbilical cord blood-derived HSPCs following cytokine stimulation. It is unknown whether AHR also contributes earlier in human hematopoietic development. To model hematopoiesis, human embryonic stem cells (hESCs) were allowed to differentiate in defined conditions in the presence of the AHR antagonist StemReginin-1 (SR-1) or the AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). We demonstrate a significant increase in CD34+CD31+ hematoendothelial cells in SR-1-treated hESCs, as well as a twofold expansion of CD34+CD45+ hematopoietic progenitor cells. Hematopoietic progenitor cells were also significantly increased by SR-1 as quantified by standard hematopoietic colony-forming assays. Using a clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-engineered hESC-RUNX1c-tdTomato reporter cell line with AHR deletion, we further demonstrate a marked enhancement of hematopoietic differentiation relative to wild-type hESCs. We also evaluated whether AHR antagonism could promote innate lymphoid cell differentiation from hESCs. SR-1 increased conventional natural killer (cNK) cell differentiation, whereas TCDD treatment blocked cNK development and supported group 3 innate lymphoid cell (ILC3) differentiation. Collectively, these results demonstrate that AHR regulates early human hematolymphoid cell development and may be targeted to enhance production of specific cell populations derived from human pluripotent stem cells.


Asunto(s)
Hematopoyesis , Células Madre Pluripotentes/citología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Humanos , Subgrupos Linfocitarios/citología , Receptores de Hidrocarburo de Aril/agonistas
2.
Eur J Immunol ; 44(12): 3717-28, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25229755

RESUMEN

Human blood NK cells exert strong cytotoxicity against transformed cells and produce different cytokines and chemokines with an important role in modulating immune responses. However, the nature of NK-cell function depends on NK-cell interaction with other immune cells. One type of immune cells that communicate with NK cells are 6-sulfo LacNAc DCs (slanDCs), which comprise a major subpopulation of proinflammatory human blood DCs. In this study, we investigated the molecular mechanisms by which slanDCs interact with NK cells. Our in vitro studies demonstrate that LPS-stimulated slanDCs enhance activation and function of NK cells essentially via membrane-bound TNF-α (mTNF-α). LPS stimulation upregulates expression of mTNF-α in slanDCs, and surface TNF receptor 2 (TNFR2) is upregulated on NK cells after coincubation with slanDCs. IL-12 secreted by slanDCs increases surface expression of TNFR2 in NK cells. TNFR2 signaling in NK cells leads to activation of NF-kB, a transcription factor for cytokines such as GM-CSF. GM-CSF provided by NK cells is responsible for enhancing IL-12 secretion in slanDCs. In conclusion, TNFR2 and IL-12 signaling, which support one another, enables slanDCs to enhance NK-cell function through mTNF-α, thereby regulating immune responses.


Asunto(s)
Membrana Celular/inmunología , Células Dendríticas/inmunología , Interleucina-12/inmunología , Células Asesinas Naturales/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Amino Azúcares/inmunología , Células Dendríticas/citología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Células Asesinas Naturales/citología , Lipopolisacáridos/farmacología , Masculino , FN-kappa B/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
3.
Cancer Immunol Immunother ; 64(9): 1175-84, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26036909

RESUMEN

Macrophage-activating lipopeptide-2 (MALP-2) is a potent inducer of proinflammatory cytokine secretion by macrophages, monocytes, and dendritic cells. MALP-2 was reported to be involved in natural killer (NK) cell activation and ensuing tumor rejection. However, the mechanism of MALP-2-mediated NK cell activation remained unclear. Therefore, we studied the effects of MALP-2 on cultured human NK cells. We found that MALP-2 had no direct effect on NK cells. Instead, MALP-2 acted on monocytes and triggered the release of different molecules such as interleukin (IL)-1ß, IL-6, IL-10, IL-12, IL-15, interferon gamma-induced protein (IP-10), and prostaglandin (PG)-E2. Our data show that monocyte-derived IP-10 could significantly induce NK cell cytotoxicity as long as the immunosuppression by PGE2 is specifically inhibited by cyclooxygenase (COX)-2 blockade. In summary, our results show that MALP-2-mediated stimulation of monocytes results in the production of several mediators which, depending on the prevailing conditions, affect the activity of NK cells in various ways. Hence, MALP-2 administration with concurrent blocking of COX-2 can be considered as a promising approach in MALP-2-based adjuvant tumor therapies.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/antagonistas & inhibidores , Células Asesinas Naturales/efectos de los fármacos , Lipopéptidos/farmacología , Monocitos/efectos de los fármacos , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 6/agonistas , Células Cultivadas , Inhibidores de la Ciclooxigenasa 2/inmunología , Dinoprostona/biosíntesis , Dinoprostona/inmunología , Humanos , Células Asesinas Naturales/inmunología , Lipopéptidos/inmunología , Monocitos/inmunología , Transducción de Señal
4.
Cell Commun Signal ; 12: 63, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25266361

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) are increasingly considered to be used as biological immunosuppressants in hematopoietic stem cell transplantation (HSCT). In the early reconstitution phase following HSCT, natural killer (NK) cells represent the major lymphocyte population in peripheral blood and display graft-vs-leukemia (GvL) effects. The functional interactions between NK cells and MSCs have the potential to influence the leukemia relapse rate after HSCT. Until date, MSC-NK cell interaction studies are largely focussed on bone marrow derived (BM)-MSCs. Umbilical cord derived (UC)-MSCs might be an alternative source of therapeutic MSCs. Thus, we studied the interaction of UC-MSCs with unstimulated allogeneic NK cells. RESULTS: UC-MSCs could potently suppress NK cell cytotoxicity in overnight cultures via soluble factors. The main soluble immunosuppressant was identified as prostaglandin (PG)-E2. Maximal PGE2 release involved IL-1ß priming of MSCs after close contact between the NK cells and UC-MSCs. Interestingly, blocking gamma-secretase activation alleviated the immunosuppression by controlling PGE2 production. IL-1 receptor activation and subsequent downstream signalling events were found to require gamma-secretase activity. CONCLUSION: Although the role of PGE2 in NK cell-MSC has been reported, the requirement of cell-cell contact for PGE2 induced immunosuppression remained unexplained. Our findings shed light on this puzzling observation and identify new players in the NK cell-MSC crosstalk.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Dinoprostona/metabolismo , Células Asesinas Naturales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Comunicación Celular , Degranulación de la Célula , Células Cultivadas , Pruebas Inmunológicas de Citotoxicidad , Humanos , Recién Nacido , Interleucina-1/metabolismo , Células K562 , Células Asesinas Naturales/fisiología , Células Madre Mesenquimatosas/fisiología , Fenotipo , Receptores de Interleucina-1/metabolismo , Cordón Umbilical/citología
5.
Stem Cells Transl Med ; 11(5): 513-526, 2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35349707

RESUMEN

Epigenetic modification is an important process during hematopoietic cell differentiation. Histone deacetylase (HDAC) inhibitors have previously been shown to enhance expansion of umbilical cord blood-derived hematopoietic stem cells (HSCs). However, the effect of HDAC inhibitors on pluripotent stem cells (PSCs) in this context is less understood. For years, investigators have considered PSC-derived natural killer (NK) and T-cell therapies. These "off-the-shelf" cellular therapies are now entering the clinic. However, the in vitro commitment of PSCs to the hematopoietic lineage is inefficient and represents a major bottleneck. We investigated whether HDAC inhibitors (HDACi) influence human PSC differentiation into CD34+CD45+ hematopoietic stem and progenitor cells (HSPCs), focusing on hemogenic endothelium (HE). Pluripotent stem cells cultured in the presence of HDACi showed a 2-5 times increase in HSPCs. Concurrent with this, HDACi-treated PSCs increased expression of 7 transcription factors (HOXA5, HOXA9, HOXA10, RUNX1, ERG, SPI1, and LCOR) recently shown to convert HE to HSPCs. ChIP-qPCR showed that SAHA upregulated acetylated-H3 at the promoter region of the above key genes. SAHA-treated human PSC-derived CD34+CD45+ cells showed primary engraftment in immunodeficient mice, but not serial transplantation. We further demonstrate that SAHA-derived HSPCs could differentiate into functional NK cells in vitro. The addition of SAHA is an easy and effective approach to overcoming the bottleneck in the transition from PSC to HSPCs for "off-the-shelf" cellular immunotherapy.


Asunto(s)
Hemangioblastos , Trasplante de Células Madre Hematopoyéticas , Células Madre Pluripotentes , Animales , Antígenos CD34/metabolismo , Diferenciación Celular , Células Cultivadas , Hemangioblastos/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Ratones
7.
Front Immunol ; 12: 607282, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33854497

RESUMEN

Over the past decade, immunotherapies have revolutionized the treatment of cancer. Although the success of immunotherapy is remarkable, it is still limited to a subset of patients. More than 1500 clinical trials are currently ongoing with a goal of improving the efficacy of immunotherapy through co-administration of other agents. Preclinical, small-animal models are strongly desired to increase the pace of scientific discovery, while reducing the cost of combination drug testing in humans. Human immune system (HIS) mice are highly immune-deficient mouse recipients rtpeconstituted with human hematopoietic stem cells. These HIS-mice are capable of growing human tumor cell lines and patient-derived tumor xenografts. This model allows rapid testing of multiple, immune-related therapeutics for tumors originating from unique clinical samples. Using a cord blood-derived HIS-BALB/c-Rag2nullIl2rγnullSIRPαNOD (BRGS) mouse model, we summarize our experiments testing immune checkpoint blockade combinations in these mice bearing a variety of human tumors, including breast, colorectal, pancreatic, lung, adrenocortical, melanoma and hematological malignancies. We present in-depth characterization of the kinetics and subsets of the HIS in lymph and non-lymph organs and relate these to protocol development and immune-related treatment responses. Furthermore, we compare the phenotype of the HIS in lymph tissues and tumors. We show that the immunotype and amount of tumor infiltrating leukocytes are widely-variable and that this phenotype is tumor-dependent in the HIS-BRGS model. We further present flow cytometric analyses of immune cell subsets, activation state, cytokine production and inhibitory receptor expression in peripheral lymph organs and tumors. We show that responding tumors bear human infiltrating T cells with a more inflammatory signature compared to non-responding tumors, similar to reports of "responding" patients in human immunotherapy clinical trials. Collectively these data support the use of HIS mice as a preclinical model to test combination immunotherapies for human cancers, if careful attention is taken to both protocol details and data analysis.


Asunto(s)
Modelos Animales de Enfermedad , Xenoinjertos , Sistema Inmunológico , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Animales , Quimerismo , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/etiología , Fenotipo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Sci Immunol ; 5(53)2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33219153

RESUMEN

Innate lymphoid cells (ILCs) develop from common lymphoid progenitors (CLPs), which further differentiate into the common ILC progenitor (CILP) that can give rise to both ILCs and natural killer (NK) cells. Murine ILC intermediates have recently been characterized, but the human counterparts and their developmental trajectories have not yet been identified, largely due to the lack of homologous surface receptors in both organisms. Here, we show that human CILPs (CD34+CD117+α4ß7+Lin-) acquire CD48 and CD52, which define NK progenitors (NKPs) and ILC precursors (ILCPs). Two distinct NK cell subsets were generated in vitro from CD34+CD117+α4ß7+Lin-CD48-CD52+ and CD34+CD117+α4ß7+Lin-CD48+CD52+ NKPs, respectively. Independent of NKPs, ILCPs exist in the CD34+CD117+α4ß7+Lin-CD48+CD52+ subset and give rise to ILC1s, ILC2s, and NCR+ ILC3s, whereas CD34+CD117+α4ß7+Lin-CD48+CD52- ILCPs give rise to a distinct subset of ILC3s that have lymphoid tissue inducer (LTi)-like properties. In addition, CD48-expressing CD34+CD117+α4ß7+Lin- precursors give rise to tissue-associated ILCs in vivo. We also observed that the interaction of 2B4 with CD48 induced differentiation of ILC2s, and together, these findings show that expression of CD48 by human ILCPs modulates ILC differentiation.


Asunto(s)
Antígeno CD48/metabolismo , Diferenciación Celular/inmunología , Células Asesinas Naturales/fisiología , Células Progenitoras Linfoides/fisiología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Animales , Antígeno CD52/metabolismo , Separación Celular , Células Cultivadas , Citometría de Flujo , Técnicas de Inactivación de Genes , Humanos , Inmunidad Innata , Ratones , Cultivo Primario de Células , RNA-Seq , Transducción de Señal/genética , Transducción de Señal/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Análisis de la Célula Individual , Especificidad de la Especie
9.
Sci Rep ; 10(1): 6335, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286456

RESUMEN

Numerous cell types modulate hematopoiesis through soluble and membrane bound molecules. Whether developing hematopoietic progenitors of a particular lineage modulate the differentiation of other hematopoietic lineages is largely unknown. Here we aimed to investigate the influence of myeloid progenitors on CD34+ cell differentiation into CD56+ innate lymphocytes. Sorted CD34+ cells cultured in the presence of stem cell factor (SCF) and FMS-like tyrosine kinase 3 ligand (FLT3L) give rise to numerous cell types, including progenitors that expressed the prolactin receptor (PRLR). These CD34+PRLR+ myeloid-lineage progenitors were derived from granulocyte monocyte precursors (GMPs) and could develop into granulocytes in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) in vitro. Moreover, CD34+PRLR+ myeloid progenitors lacked lymphoid developmental potential, but when stimulated with prolactin (PRL) they increased the differentiation of other CD34+ cell populations into the NK lineage in a non-contact dependent manner. Both mRNA and protein analyses show that PRL increased mothers against decapentaplegic homolog 7 (SMAD7) in CD34+PRLR+ myeloid cells, which reduced the production of transforming growth factor beta 1 (TGF-ß1), a cytokine known to inhibit CD56+ cell development. Thus, we uncover an axis whereby CD34+PRLR+ GMPs inhibit CD56+ lineage development through TGF-ß1 production and PRL stimulation leads to SMAD7 activation, repression of TGF-ß1, resulting in CD56+ cell development.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Linfopoyesis/genética , Prolactina/genética , Receptores de Prolactina/genética , Proteína smad7/genética , Factor de Crecimiento Transformador beta1/genética , Antígenos CD34/genética , Antígenos CD34/inmunología , Antígeno CD56/genética , Antígeno CD56/inmunología , Diferenciación Celular/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Linfocitos/citología , Linfocitos/inmunología , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Tirosina Quinasa 3 Similar a fms/genética
10.
Front Immunol ; 10: 510, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30949172

RESUMEN

Helper Innate lymphoid cells (ILCs) are tissue resident lymphocytes that play a critical role in a number of biological processes. Several transcription factors are required for the differentiation of hematopoietic stem cells (HSCs) into ILCs. Recent studies demonstrate GATA3 as a transcriptional regulator that plays an essential role in ILC development. We aimed to modulate the differentiation of human cord blood-derived CD34+ cells into ILCs by transient and ectopic expression of mRNA encoding transcription factors known to be important for ILC lineage differentiation, including GATA3, TOX, NFIL3, ID2, and RORγt. Using this experimental protocol, only GATA3 significantly modulated HSCs to differentiate into helper ILCs. Transient overexpression of GATA3 drove the emergence of CD34+α4ß7+ early ILC progenitors during the first few days of culture. These ILC progenitors further acquired IL-7Rα and CD117 to give rise to immediate ILC precursors. In support of these findings, analysis of the genes induced by GATA3 in HSCs showed an upregulation of those associated with ILC development. Moreover, we show GATA3 also acts on more committed progenitors and significantly shifts the differentiation of progenitors away from the ILC1/NK lineage to the ILC2 and ILC3 lineage. In summary, transient overexpression of GATA3 mRNA in CD34+ HSCs enhances the differentiation of HSCs into the helper ILC lineages, at the expense of NK cell development.


Asunto(s)
Diferenciación Celular/inmunología , Factor de Transcripción GATA3/inmunología , Regulación de la Expresión Génica/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunidad Innata , Linfocitos T Colaboradores-Inductores/inmunología , Células Madre Hematopoyéticas/citología , Humanos , Linfocitos T Colaboradores-Inductores/citología
11.
Cell Mol Immunol ; 14(12): 976-985, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27086951

RESUMEN

To function optimally, human blood natural killer (NK) cells need to communicate with other immune cells. Previously, it has been shown that NK cells communicate with 6-sulfo LacNAc dendritic cells (slanDCs), which are able to stimulate NK cells in vitro. In this study, we investigated how slanDCs regulate the level of NK cell activation. The secretion of interleukin (IL)-1ß by slanDCs during coculture with NK cells increased as a result of signaling via intercellular adhesion molecule-1 on slanDCs following its interaction with lymphocyte function-associated antigen-1 on NK cells. IL-1ß induced the expression of Fas receptor (CD95) on NK cells. The binding of Fas ligand (CD178) to CD95 induced the apoptosis of activated NK cells. Moreover, IL-1ß also induced increased cyclooxygenase-2 expression in slanDCs, which in turn enabled the cells to secrete prostaglandin (PG)-E2. Consequently, PGE2 acted as a suppressing agent, tuning down the activation level of NK cells. In summary, IL-1ß limits the level of NK cell activation by inducing apoptosis and suppression as a homeostatic regulatory function.


Asunto(s)
Células Dendríticas/inmunología , Inflamación/inmunología , Interleucina-1beta/metabolismo , Células Asesinas Naturales/inmunología , Receptor fas/metabolismo , Amino Azúcares/metabolismo , Apoptosis , Comunicación Celular , Células Cultivadas , Técnicas de Cocultivo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Proteína Ligando Fas/metabolismo , Regulación de la Expresión Génica , Humanos , Terapia de Inmunosupresión , Activación de Linfocitos , Receptor fas/genética
12.
J Acquir Immune Defic Syndr ; 73(1): 34-8, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27243902

RESUMEN

HIV infection is marked by phenotypic and functional alterations of immune cells. Different studies have shown both numerical and functional deterioration of dendritic cells in HIV-1-infected patients. In this study, we report an increase of inflammatory 6-sulfo LacNAc dendritic cells (slanDCs) that are more activated and produce higher amounts of interleukin (IL)-1ß during HIV-1 infection as compared with healthy controls. IL-1ß plays a regulatory role in chronic inflammatory disorders. Therefore, our findings might reveal a compensatory regulatory function of slanDCs during HIV-1 infection.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por VIH/inmunología , Interleucina-1beta/biosíntesis , Adulto , Recuento de Linfocito CD4 , Estudios de Casos y Controles , Femenino , VIH-1/genética , Humanos , Masculino , Persona de Mediana Edad , Carga Viral
13.
Cell Rep ; 17(4): 1113-1127, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760315

RESUMEN

Cytomegalovirus (CMV) is an opportunistic virus severely infecting immunocompromised individuals. In mice, endosomal Toll-like receptor 9 (TLR9) and downstream myeloid differentiation factor 88 (MyD88) are central to activating innate immune responses against mouse CMV (MCMV). In this respect, the cell-specific contribution of these pathways in initiating anti-MCMV immunity remains unclear. Using transgenic mice, we demonstrate that TLR9/MyD88 signaling selectively in CD11c+ dendritic cells (DCs) strongly enhances MCMV clearance by boosting natural killer (NK) cell CD69 expression and IFN-γ production. In addition, we show that in the absence of plasmacytoid DCs (pDCs), conventional DCs (cDCs) promote robust NK cell effector function and MCMV clearance in a TLR9/MyD88-dependent manner. Simultaneously, cDC-derived IL-15 regulates NK cell degranulation by TLR9/MyD88-independent mechanisms. Overall, we compartmentalize the cellular contribution of TLR9 and MyD88 signaling in individual DC subsets and evaluate the mechanism by which cDCs control MCMV immunity.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Infecciones por Citomegalovirus/virología , Células Dendríticas/metabolismo , Muromegalovirus/fisiología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Receptor Toll-Like 9/metabolismo , Animales , Antivirales/farmacología , Antígeno CD11c/metabolismo , Citotoxicidad Inmunológica , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Ratones Endogámicos BALB C
14.
AIDS Res Hum Retroviruses ; 31(12): 1206-12, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26352913

RESUMEN

HIV-1 infection results in immunological abnormalities of natural killer (NK) cells such as disturbed distribution of NK cell subsets and downmodulation of activating and upregulation of inhibitory receptors thereby diminishing NK cell killing capacity and cytokine secretion. Antiretroviral treatment (ART) is known to restore phenotype and functions of NK cells. However, the effects of ART on NK cell terminal differentiation, activation, and disturbed distribution have not been studied yet longitudinally. Here, we analyzed the effects of ART on these parameters of peripheral blood NK cells in a longitudinal as well as in a cross-sectional study. We observed that expanded CD56(-)CD16(+) NK cell frequency is inversely correlated with the frequency of CD56(dim)CD16(+) NK cells in treatment-naive HIV-1 patients. Loss of CD56(dim)CD16(+) and expansion of CD56(-)CD16(+) NK cells again restore to the levels of healthy controls after ART. Enhanced immune activation of different NK cell subsets is partially restored after ART. Terminal differentiation of CD56(dim)CD16(+) NK cells is enhanced after ART as measured by CD57 expression. Frequencies of CD57(+)CD56(dim)CD16(+) NK cells are directly correlated with the frequencies of total NK cells suggesting that an increase in the frequencies of CD57(+)CD56(dim)CD16(+) NK cells is reflected by increased frequencies of total NK cells after ART. Taken together these data demonstrate that ART has an effect on the immune restoration of NK cells and is enhanced in the terminal differentiation of CD56(dim)CD16(+) NK cells, which is associated with increased frequencies of total NK cells after ART.


Asunto(s)
Antirretrovirales/uso terapéutico , Diferenciación Celular , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/fisiología , Antígeno CD56/análisis , Antígenos CD57/análisis , Estudios Transversales , Proteínas Ligadas a GPI/análisis , Infecciones por VIH/virología , VIH-1/inmunología , Humanos , Células Asesinas Naturales/química , Estudios Longitudinales , Subgrupos Linfocitarios/química , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/fisiología , Receptores de IgG/análisis
15.
Front Immunol ; 5: 662, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25584044

RESUMEN

Following allogeneic hematopoietic stem cell transplantation (HSCT), interferon (IFN)-γ levels in the recipient's body can strongly influence the clinical outcome. Human umbilical cord-derived mesenchymal stem cells (UC-MSCs) are lucrative as biological tolerance-inducers in HSCT settings. Hence, we studied the molecular mechanism of how UC-MSCs influence natural killer (NK) cell-mediated IFN-γ production. Allogeneic NK cells were cultured in direct contact with UC-MSCs or cell-free supernatants from mesenchymal stem cell (MSC) cultures (MSC-conditioned media). We found that soluble factors secreted by UC-MSCs strongly suppressed interleukin (IL)-12/IL-18-induced IFN-γ production by NK cells by reducing phosphorylation of STAT4, NF-κB, as well as T-bet activity. UC-MSCs secreted considerable amounts of activin-A, which could suppress IFN-γ production by NK cells. Neutralization of activin-A in MSC-conditioned media significantly abrogated their suppressive abilities. Till date, multiple groups have reported that prostaglandin (PG)-E2 produced by MSCs can suppress NK cell functions. Indeed, we found that inhibition of PGE2 production by MSCs could also significantly restore IFN-γ production. However, the effects of activin-A and PGE2 were not cumulative. To the best of our knowledge, we are first to report the role of activin-A in MSC-mediated suppression of IFN-γ production by NK cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA