Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Microbiol ; 23(1): 394, 2023 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-38066426

RESUMEN

Intrinsic metabolism shapes the immune environment associated with immune suppression and tolerance in settings such as organ transplantation and cancer. However, little is known about the metabolic activities in an immunosuppressive environment. In this study, we employed metagenomic, metabolomic, and immunological approaches to profile the early effects of the immunosuppressant drug tacrolimus, antibiotics, or both in gut lumen and circulation using a murine model. Tacrolimus induced rapid and profound alterations in metabolic activities within two days of treatment, prior to alterations in gut microbiota composition and structure. The metabolic profile and gut microbiome after seven days of treatment was distinct from that after two days of treatment, indicating continuous drug effects on both gut microbial ecosystem and host metabolism. The most affected taxonomic groups are Clostriales and Verrucomicrobiae (i.e., Akkermansia muciniphila), and the most affected metabolic pathways included a group of interconnected amino acids, bile acid conjugation, glucose homeostasis, and energy production. Highly correlated metabolic changes were observed between lumen and serum metabolism, supporting their significant interactions. Despite a small sample size, this study explored the largely uncharacterized microbial and metabolic events in an immunosuppressed environment and demonstrated that early changes in metabolic activities can have significant implications that may serve as antecedent biomarkers of immune activation or quiescence. To understand the intricate relationships among gut microbiome, metabolic activities, and immune cells in an immune suppressed environment is a prerequisite for developing strategies to monitor and optimize alloimmune responses that determine transplant outcomes.


Asunto(s)
Tacrolimus , Animales , Ratones , Inmunosupresores/farmacología , Metaboloma , Metabolómica
2.
Am J Transplant ; 20(9): 2343-2355, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32282980

RESUMEN

Myeloid-derived suppressor cells (MDSCs) expand in an inflammatory microenvironment such as cancer and autoimmunity. To study if transplantation induces MDSCs and these cells regulate allograft survival, C57BL/6 donor hearts were transplanted into BALB/c recipients and endogenous MDSCs were characterized. The effects of adoptive transfer of transplant (tx), tumor (tm), and granulocyte-colony stimulating factor (g-csf)-expanded MDSCs or depletion of MDSC were assessed. MDSCs expanded after transplantation (1.7-4.6-fold) in the absence of immunosuppression, homed to allografts, and suppressed proliferation of CD4 T cells in vitro. Tx-MDSCs differed phenotypically from tm-MDSCs and g-csf-MDSCs. Among various surface markers, Rae-1 expression was notably low and TGF-ß receptor II was high in tx-MDSCs when compared to tm-MDSCs and g-csf-MDSCs. Adoptive transfer of these three MDSCs led to differential graft survival: control (6 days), tx-MDSCs (7.5 days), tm-MDSCs (9.5 days), and g-csf-MDSCs (19.5 days). In combination with anti-CD154 mAb, MDSCs synergistically extended graft survival from 40 days (anti-CD154 alone) to 86 days with tm-MDSCs and 132 days with g-csf-MDSCs. Early MDSC depletion (day 0 or 20), however, abrogated graft survival, but late depletion (day 25) did not. In conclusion, MDSCs expanded following transplantation, migrated to cardiac allografts, prolonged graft survival, and were synergistic with anti-CD154 mAb.


Asunto(s)
Trasplante de Corazón , Células Supresoras de Origen Mieloide , Animales , Supervivencia de Injerto , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Donantes de Tejidos
3.
Proc Natl Acad Sci U S A ; 110(47): 19036-41, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24194546

RESUMEN

Toll/IL-1R resistance (TIR) domain-containing adapter-inducing IFN-ß (TRIF) is a Toll-like receptor (TLR) adapter that mediates MyD88-independent induction of type I interferons through activation of IFN regulatory factor 3 and NFκB. We have examined peptides derived from the TRIF TIR domain for ability to inhibit TLR4. In addition to a previously identified BB loop peptide (TF4), a peptide derived from putative helix B of TRIF TIR (TF5) strongly inhibits LPS-induced cytokine and MAPK activation in wild-type cells. TF5 failed to inhibit LPS-induced cytokine and kinase activation in TRIF-deficient immortalized bone-marrow-derived macrophage, but was fully inhibitory in MyD88 knockout cells. TF5 does not block macrophage activation induced by TLR2, TLR3, TLR9, or retinoic acid-inducible gene 1/melanoma differentiation-associated protein 5 agonists. Immunoprecipitation assays demonstrated that TF4 binds to TLR4 but not TRIF-related adaptor molecule (TRAM), whereas TF5 binds to TRAM strongly and TLR4 to a lesser extent. Although TF5 prevented coimmunoprecipitation of TRIF with both TRAM and TLR4, site-directed mutagenesis of the TRIF B helix residues affected TRIF-TRAM coimmunoprecipitation selectively, as these mutations did not block TRIF-TLR4 association. These results suggest that the folded TRIF TIR domain associates with TRAM through the TRIF B helix region, but uses a different region for TRIF-TLR4 association. The B helix peptide TF5, however, can associate with either TRAM or TLR4. In a mouse model of TLR4-driven inflammation, TF5 decreased plasma cytokine levels and protected mice from a lethal LPS challenge. Our data identify TRIF sites that are important for interaction with TLR4 and TRAM, and demonstrate that TF5 is a potent TLR4 inhibitor with significant potential as a candidate therapeutic for human sepsis.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Interferón beta/metabolismo , Receptores de Interleucina/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 4/metabolismo , Animales , Dicroismo Circular , Escherichia coli , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida
4.
J Immunol ; 190(5): 2263-72, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23345333

RESUMEN

Toll/IL-1R (TIR) domain-containing adapter-inducing IFN-ß (TRIF)-related adapter molecule (TRAM) serves as a bridging adapter that enables recruitment of TRIF to activated TLR4 and thereby mediates the induction of TRIF-dependent cytokines. A library of cell-permeating decoy peptides derived from TRAM TIR domain has been screened for the ability of individual peptides to inhibit TLR4 signaling in primary murine macrophages. Peptides derived from TRAM TIR BB loop (TM4) and C helix (TM6) inhibited the LPS-induced activation of MyD88-dependent and TRIF-dependent cytokines, as well as MAPK activation. TM4 and TM6 did not block macrophage activation induced by TLR2, TLR9, or retinoic acid-inducible gene 1-like receptor agonists. Both TM4 and TM6 blocked coimmunoprecipitation of TRAM and TLR4 ectopically expressed in HEK293T cells. Both peptides also blocked the LPS-induced recruitment of MyD88 to TLR4 in primary murine macrophages. In vivo examination of TRAM-derived peptides demonstrated that all peptides that were inhibitory in vitro profoundly suppressed systemic inflammatory response elicited in mice by a sublethal LPS dose, and protected mice against a lethal LPS challenge. This research identifies novel TLR inhibitors effective in vitro and in vivo and validates the approach taken in this study as a rational way for development of signaling inhibitors and lead therapeutics.


Asunto(s)
Inflamación/tratamiento farmacológico , Macrófagos Peritoneales/efectos de los fármacos , Péptidos/farmacología , Receptores de Interleucina/genética , Receptor Toll-Like 4/antagonistas & inhibidores , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Secuencia de Aminoácidos , Animales , Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Lipopolisacáridos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/inmunología , Datos de Secuencia Molecular , Factor 88 de Diferenciación Mieloide/antagonistas & inhibidores , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Biblioteca de Péptidos , Péptidos/síntesis química , Cultivo Primario de Células , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Interleucina/química , Receptores de Interleucina/inmunología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
5.
J Immunol ; 188(9): 4506-15, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22474023

RESUMEN

Asp(299)Gly (D299G) and, to a lesser extent, Thr(399)Ile (T399I) TLR4 polymorphisms have been associated with gram-negative sepsis and other infectious diseases, but the mechanisms by which they affect TLR4 signaling are unclear. In this study, we determined the impact of the D299G and T399I polymorphisms on TLR4 expression, interactions with myeloid differentiation factor 2 (MD2), LPS binding, and LPS-mediated activation of the MyD88- and Toll/IL-1R resistance domain-containing adapter inducing IFN-ß (TRIF) signaling pathways. Complementation of human embryonic kidney 293/CD14/MD2 transfectants with wild-type (WT) or mutant yellow fluorescent protein-tagged TLR4 variants revealed comparable total TLR4 expression, TLR4-MD2 interactions, and LPS binding. FACS analyses with anti-TLR4 Ab showed only minimal changes in the cell-surface levels of the D299G TLR4. Cells transfected with D299G TLR4 exhibited impaired LPS-induced phosphorylation of p38 and TANK-binding kinase 1, activation of NF-κB and IFN regulatory factor 3, and induction of IL-8 and IFN-ß mRNA, whereas T399I TLR4 did not cause statistically significant inhibition. In contrast to WT TLR4, expression of the D299G mutants in TLR4(-/-) mouse macrophages failed to elicit LPS-mediated induction of TNF-α and IFN-ß mRNA. Coimmunoprecipitation revealed diminished LPS-driven interaction of MyD88 and TRIF with the D299G TLR4 species, in contrast to robust adapter recruitment exhibited by WT TLR4. Thus, the D299G polymorphism compromises recruitment of MyD88 and TRIF to TLR4 without affecting TLR4 expression, TLR4-MD2 interaction, or LPS binding, suggesting that it interferes with TLR4 dimerization and assembly of intracellular docking platforms for adapter recruitment.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Macrófagos/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Polimorfismo Genético/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/genética , Antígeno 96 de los Linfocitos/inmunología , Antígeno 96 de los Linfocitos/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Fosforilación/inmunología , Proteínas Quinasas/genética , Proteínas Quinasas/inmunología , Proteínas Quinasas/metabolismo , Multimerización de Proteína/genética , Multimerización de Proteína/inmunología , Transducción de Señal/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
6.
Transplantation ; 108(7): e91-e105, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38587506

RESUMEN

BACKGROUND: Despite ongoing improvements to regimens preventing allograft rejection, most cardiac and other organ grafts eventually succumb to chronic vasculopathy, interstitial fibrosis, or endothelial changes, and eventually graft failure. The events leading to chronic rejection are still poorly understood and the gut microbiota is a known driving force in immune dysfunction. We previously showed that gut microbiota dysbiosis profoundly influences the outcome of vascularized cardiac allografts and subsequently identified biomarker species associated with these differential graft outcomes. METHODS: In this study, we further detailed the multifaceted immunomodulatory properties of protolerogenic and proinflammatory bacterial species over time, using our clinically relevant model of allogenic heart transplantation. RESULTS: In addition to tracing longitudinal changes in the recipient gut microbiome over time, we observed that Bifidobacterium pseudolongum induced an early anti-inflammatory phenotype within 7 d, whereas Desulfovibrio desulfuricans resulted in a proinflammatory phenotype, defined by alterations in leukocyte distribution and lymph node (LN) structure. Indeed, in vitro results showed that B pseudolongum and D desulfuricans acted directly on primary innate immune cells. However, by 40 d after treatment, these 2 bacterial strains were associated with mixed effects in their impact on LN architecture and immune cell composition and loss of colonization within gut microbiota, despite protection of allografts from inflammation with B pseudolongum treatment. CONCLUSIONS: These dynamic effects suggest a critical role for early microbiota-triggered immunologic events such as innate immune cell engagement, T-cell differentiation, and LN architectural changes in the subsequent modulation of protolerant versus proinflammatory immune responses in organ transplant recipients.


Asunto(s)
Bifidobacterium , Microbioma Gastrointestinal , Rechazo de Injerto , Trasplante de Corazón , Trasplante de Corazón/efectos adversos , Microbioma Gastrointestinal/inmunología , Rechazo de Injerto/inmunología , Rechazo de Injerto/microbiología , Rechazo de Injerto/prevención & control , Animales , Masculino , Factores de Tiempo , Supervivencia de Injerto , Disbiosis , Ratones Endogámicos C57BL , Inmunidad Innata , Inmunomodulación , Fenotipo , Probióticos/uso terapéutico , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/inmunología
7.
J Biol Chem ; 287(29): 24641-8, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22648407

RESUMEN

Toll/interleukin-1 receptor (TIR) domain-containing adapter protein/MyD88 adapter-like (TIRAP/Mal) is an adapter protein that facilitates recruitment of MyD88 to TLR4 and TLR2 signaling complexes. We previously generated a library of cell-permeating TLR4 TIR-derived decoy peptides fused to the translocating segment of the Drosophila Antennapedia homeodomain and examined each peptide for the ability to inhibit TLR4 signaling (Toshchakov, V. Y., Szmacinski, H., Couture, L. A., Lakowicz, J. R., and Vogel, S. N. (2011) J. Immunol. 186, 4819-4827). We have now expanded this study to test TIRAP decoy peptides. Five TIRAP peptides, TR3 (for TIRAP region 3), TR5, TR6, TR9, and TR11, inhibited LPS-induced cytokine mRNA expression and MAPK activation. Inhibition was confirmed at the protein level; select peptides abolished the LPS-induced cytokine production measured in cell culture 24 h after a single treatment. Two of the TLR4 inhibitory peptides, TR3 and TR6, also inhibited cytokine production induced by a TLR2/TLR1 agonist, S-(2,3-bis(palmitoyloxy)-(2R,2S)-propyl)-N-palmitoyl-(R)-Cys-Ser-Lys(4)-OH; however, a higher peptide concentration was required to achieve comparable inhibition of TLR2 versus TLR4 signaling. Two TLR4 inhibitory peptides, TR5 and TR6, were examined for the ability to inhibit TLR4-driven cytokine induction in mice. Pretreatment with either peptide significantly reduced circulating TNF-α and IL-6 in mice following LPS injection. This study has identified novel TLR inhibitory peptides that block cellular signaling at low micromolar concentrations in vitro and in vivo. Comparison of TLR4 inhibition by TLR4 and TIRAP TIR-derived peptides supports the view that structurally diverse regions mediate functional interactions of TIR domains.


Asunto(s)
Glicoproteínas de Membrana/química , Péptidos/química , Péptidos/farmacología , Receptores de Interleucina-1/química , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/metabolismo , Animales , Western Blotting , Citocinas/genética , Electroforesis en Gel de Poliacrilamida , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptores Toll-Like/agonistas , Receptores Toll-Like/antagonistas & inhibidores , Receptores Toll-Like/genética
8.
Sci Rep ; 13(1): 1023, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36658194

RESUMEN

The beneficial effects attributed to Bifidobacterium are largely attributed to their immunomodulatory capabilities, which are likely to be species- and even strain-specific. However, their strain-specificity in direct and indirect immune modulation remain largely uncharacterized. We have shown that B. pseudolongum UMB-MBP-01, a murine isolate strain, is capable of suppressing inflammation and reducing fibrosis in vivo. To ascertain the mechanism driving this activity and to determine if it is specific to UMB-MBP-01, we compared it to a porcine tropic strain B. pseudolongum ATCC25526 using a combination of cell culture and in vivo experimentation and comparative genomics approaches. Despite many shared features, we demonstrate that these two strains possess distinct genetic repertoires in carbohydrate assimilation, differential activation signatures and cytokine responses signatures in innate immune cells, and differential effects on lymph node morphology with unique local and systemic leukocyte distribution. Importantly, the administration of each B. pseudolongum strain resulted in major divergence in the structure, composition, and function of gut microbiota. This was accompanied by markedly different changes in intestinal transcriptional activities, suggesting strain-specific modulation of the endogenous gut microbiota as a key to immune modulatory host responses. Our study demonstrated a single probiotic strain can influence local, regional, and systemic immunity through both innate and adaptive pathways in a strain-specific manner. It highlights the importance to investigate both the endogenous gut microbiome and the intestinal responses in response to probiotic supplementation, which underpins the mechanisms through which the probiotic strains drive the strain-specific effect to impact health outcomes.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Ratones , Animales , Porcinos , Bifidobacterium , Intestinos , Inmunidad
9.
JCI Insight ; 8(8)2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-37092548

RESUMEN

Fibroblastic reticular cells (FRCs) play important roles in tolerance by producing laminin α4 (Lama4) and altering lymph node (LN) structure and function. The present study revealed the specific roles of extracellular matrix Lama4 in regulating LN conduits using FRC-specific KO mouse strains. FRC-derived Lama4 maintained conduit fiber integrity, as its depletion altered conduit morphology and structure and reduced homeostatic conduit flow. Lama4 regulated the lymphotoxin ß receptor (LTßR) pathway, which is critical for conduit and LN integrity. Depleting LTßR in FRCs further reduced conduits and impaired reticular fibers. Lama4 was indispensable for FRC generation and survival, as FRCs lacking Lama4 displayed reduced proliferation but upregulated senescence and apoptosis. During acute immunization, FRC Lama4 deficiency increased antigen flow through conduits. Importantly, adoptive transfer of WT FRCs to FRC Lama4-deficient mice rescued conduit structure, ameliorated Treg and chemokine distribution, and restored transplant allograft acceptance, which were all impaired by FRC Lama4 depletion. Single-cell RNA sequencing analysis of LN stromal cells indicated that the laminin and collagen signaling pathways linked crosstalk among FRC subsets and endothelial cells. This study demonstrated that FRC Lama4 is responsible for maintaining conduits by FRCs and can be harnessed to potentiate FRC-based immunomodulation.


Asunto(s)
Células Endoteliales , Laminina , Ratones , Animales , Laminina/genética , Laminina/metabolismo , Ganglios Linfáticos , Transducción de Señal , Quimiocinas/metabolismo
10.
Res Sq ; 2023 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-37790403

RESUMEN

Intrinsic metabolism shapes the immune environment associated with immune suppression and tolerance in settings such as organ transplantation and cancer. However, little is known about the metabolic activities in an immunosuppressive environment. In this study, we employed metagenomic, metabolomic, and immunological approaches to profile the early effects of the immunosuppressant drug tacrolimus, antibiotics, or both in gut lumen and circulation using a murine model. Tacrolimus induced rapid and profound alterations in metabolic activities within two days of treatment, prior to alterations in gut microbiota composition and structure. The metabolic profile and gut microbiome after seven days of treatment was distinct from that after two days of treatment, indicating continuous drug effects on both gut microbial ecosystem and host metabolism. The most affected taxonomic groups are Clostriales and Verrucomicrobiae (i.e., Akkermansia muciniphila), and the most affected metabolic pathways included a group of interconnected amino acids, bile acid conjugation, glucose homeostasis, and energy production. Highly correlated metabolic changes were observed between lumen and serum metabolism, supporting their significant interactions. Despite a small sample size, this study explored the largely uncharacterized microbial and metabolic events in an immunosuppressed environment and demonstrated that early changes in metabolic activities can have significant implications that may serve as antecedent biomarkers of immune activation or quiescence. To understand the intricate relationships among gut microbiome, metabolic activities, and immune cells in an immune suppressed environment is a prerequisite for developing strategies to monitor and optimize alloimmune responses that determine transplant outcomes.

11.
J Biol Chem ; 286(10): 7905-7916, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21220427

RESUMEN

Endotoxin tolerance reprograms Toll-like receptor 4 responses by impairing LPS-elicited production of pro-inflammatory cytokines without inhibiting expression of anti-inflammatory or anti-microbial mediators. In septic patients, Toll-like receptor tolerance is thought to underlie decreased pro-inflammatory cytokine expression in response to LPS and increased incidence of microbial infections. The impact of endotoxin tolerance on recruitment, post-translational modifications and signalosome assembly of IL-1 receptor-associated kinase (IRAK) 4, IRAK1, TNF receptor-associated factor (TRAF) 6, TGF-ß-activated kinase (TAK) 1, and IκB kinase (IKK) γ is largely unknown. We report that endotoxin tolerization of THP1 cells and human monocytes impairs LPS-mediated receptor recruitment and activation of IRAK4, ablates K63-linked polyubiquitination of IRAK1 and TRAF6, compromises assembly of IRAK1-TRAF6 and IRAK1-IKKγ platforms, and inhibits TAK1 activation. Deficiencies in these signaling events in LPS-tolerant cells coincided with increased expression of A20, an essential deubiquitination enzyme, and sustained A20-IRAK1 associations. Overexpression of A20 inhibited LPS-induced activation of NF-κB and ablated NF-κB reporter activation driven by ectopic expression of MyD88, IRAK1, IRAK2, TRAF6, and TAK1/TAB1, while not affecting the responses induced by IKKß and p65. A20 shRNA knockdown abolished LPS tolerization of THP1 cells, mechanistically linking A20 and endotoxin tolerance. Thus, deficient LPS-induced activation of IRAK4 and TAK1, K63-linked polyubiquitination of IRAK1 and TRAF6, and disrupted IRAK1-TRAF6 and IRAK1-IKKγ assembly associated with increased A20 expression and A20-IRAK1 interactions are new determinants of endotoxin tolerance.


Asunto(s)
Resistencia a Medicamentos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Lipopolisacáridos/farmacología , Quinasas Quinasa Quinasa PAM/metabolismo , Monocitos/metabolismo , Proteínas Nucleares/biosíntesis , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitinación/efectos de los fármacos , Línea Celular , Proteínas de Unión al ADN , Humanos , Quinasa I-kappa B/genética , Quinasas Asociadas a Receptores de Interleucina-1/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Quinasa Quinasa PAM/genética , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas Nucleares/genética , Factor 6 Asociado a Receptor de TNF/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
12.
Cell Rep ; 39(3): 110727, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35443187

RESUMEN

Regulatory T cell (Treg) lymphatic migration is required for resolving inflammation and prolonging allograft survival. Focusing on Treg interactions with lymphatic endothelial cells (LECs), we dissect mechanisms and functional consequences of Treg transendothelial migration (TEM). Using three genetic mouse models of pancreatic islet transplantation, we show that Treg lymphotoxin (LT) αß and LEC LTß receptor (LTßR) signaling are required for efficient Treg migration and suppressive function to prolong allograft survival. Inhibition of LT signaling increases Treg conversion to Foxp3loCD25lo exTregs. In a transwell-based model of TEM across polarized LECs, non-migrated Tregs become exTregs. Such conversion is regulated by LTßR nuclear factor κB (NF-κB) signaling in LECs, which increases interleukin-6 (IL-6) production and drives exTreg conversion. Migrating Tregs are ectonucleotidase CD39hi and resist exTreg conversion in an adenosine-receptor-2A-dependent fashion. Human Tregs migrating across human LECs behave similarly. These molecular interactions can be targeted for therapeutic manipulation of immunity and suppression.


Asunto(s)
Células Endoteliales , Linfocitos T Reguladores , Adenosina , Animales , Factores de Transcripción Forkhead/genética , Linfotoxina beta , Ratones , FN-kappa B
13.
Nat Commun ; 13(1): 2176, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35449134

RESUMEN

Programmed death-1 (PD-1) and its ligand PD-L1 are checkpoint molecules which regulate immune responses. Little is known about their functions in T cell migration and there are contradictory data about their roles in regulatory T cell (Treg) function. Here we show activated Tregs and CD4 effector T cells (Teffs) use PD-1/PD-L1 and CD80/PD-L1, respectively, to regulate transendothelial migration across lymphatic endothelial cells (LECs). Antibody blockade of Treg PD-1, Teff CD80 (the alternative ligand for PD-L1), or LEC PD-L1 impairs Treg or Teff migration in vitro and in vivo. PD-1/PD-L1 signals through PI3K/Akt and ERK to regulate zipper junctional VE-cadherin, and through NFκB-p65 to up-regulate VCAM-1 expression on LECs. CD80/PD-L1 signaling up-regulates VCAM-1 through ERK and NFκB-p65. PD-1 and CD80 blockade reduces tumor egress of PD-1high fragile Tregs and Teffs into draining lymph nodes, respectively, and promotes tumor regression. These data provide roles for PD-L1 in cell migration and immune regulation.


Asunto(s)
Antígeno B7-H1 , Receptor de Muerte Celular Programada 1 , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Células Endoteliales/metabolismo , Ligandos , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T Reguladores , Migración Transendotelial y Transepitelial , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
J Clin Invest ; 132(13)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35775481

RESUMEN

Lymph node (LN) fibroblastic reticular cells (FRCs) define LN niches and regulate lymphocyte homeostasis through producing diverse extracellular matrix (ECM) components. We examined the role of ECM laminin α4 (Lama4) using FRC-Lama4 conditional KO Pdgfrb-Cre-/- × Lama4fl/fl mice. Single-cell RNA-sequencing (scRNA-Seq) data showed the promoter gene Pdgfrb was exclusively expressed in FRCs. Depleting FRC-Lama4 reduced Tregs and dendritic cells, decreased high endothelial venules, impaired the conduit system, and downregulated T cell survival factors in LNs. FRC-Lama4 depletion impaired the homing of lymphocytes to LNs in homeostasis and after allografting. Alloantigen-specific T cells proliferated, were activated to greater degrees in LNs lacking FRC-Lama4, and were more prone to differentiate into effector phenotypes relative to the Treg phenotype. In murine cardiac transplantation, tolerogenic immunosuppression was not effective in FRC-Lama4 recipients, which produced more alloantibodies than WT. After lung transplantation, FRC-Lama4-KO mice had more severe graft rejection with fewer Tregs in their LNs. Overall, FRC-Lama4 critically contributes to a tolerogenic LN niche by supporting T cell migration, constraining T cell activation and proliferation, and promoting Treg differentiation. Hence, it serves as a therapeutic target for immunoengineering.


Asunto(s)
Laminina , Ganglios Linfáticos , Reticulina , Linfocitos T , Animales , Laminina/inmunología , Ganglios Linfáticos/inmunología , Ratones , Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Reticulina/inmunología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Inmunología del Trasplante
15.
Cells ; 10(4)2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33805271

RESUMEN

The pleiotropic functions of lymphotoxin (LT)ß receptor (LTßR) signaling are linked to the control of secondary lymphoid organ development and structural maintenance, inflammatory or autoimmune disorders, and carcinogenesis. Recently, LTßR signaling in endothelial cells has been revealed to regulate immune cell migration. Signaling through LTßR is comprised of both the canonical and non-canonical-nuclear factor κB (NF-κB) pathways, which induce chemokines, cytokines, and cell adhesion molecules. Here, we focus on the novel functions of LTßR signaling in lymphatic endothelial cells for migration of regulatory T cells (Tregs), and specific targeting of LTßR signaling for potential therapeutics in transplantation and cancer patient survival.


Asunto(s)
Movimiento Celular , Leucocitos/citología , Leucocitos/metabolismo , Sistema Linfático/citología , Receptor beta de Linfotoxina/metabolismo , Transducción de Señal , Células Endoteliales/metabolismo , Humanos
16.
Cell Rep ; 30(4): 1052-1062.e5, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31995749

RESUMEN

Regulatory T cells (Tregs) express high levels of cell surface lymphotoxin alpha beta (LTα1ß2) to activate the LT beta receptor (LTßR) on the lymphatic endothelial cells (LECs), modulating LEC adhesion molecules, intercellular junctions, and chemokines. We demonstrate a role for Tregs through this pathway to condition the permissiveness of lymphatic endothelia for transendothelial migration (TEM), thus gating leukocyte traffic. Human Tregs share the same property with murine Tregs. Activation of TLR2 on Tregs during inflammation specifically augments LTα1ß2-LTßR signaling, which further enhances the permissiveness of LECs to facilitate TEM. The conditioning of endothelia may promote the resolution of inflammation by directing leukocytes out of tissues to lymphatic vessels and draining lymph nodes (dLNs). Thus, Tregs interact with lymphatic endothelia under homeostasis and inflammation and dictate endothelial permissiveness and gating mechanisms for subsequent leukocyte migration through endothelial barriers.


Asunto(s)
Movimiento Celular/inmunología , Endotelio Linfático/metabolismo , Inflamación/metabolismo , Linfocitos T Reguladores/metabolismo , Receptor Toll-Like 2/metabolismo , Migración Transendotelial y Transepitelial/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Cadherinas/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Quimiocina CCL21/metabolismo , Endotelio Linfático/efectos de los fármacos , Humanos , Inflamación/inmunología , Inflamación/patología , Interleucina-2/farmacología , Islotes Pancreáticos/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Receptor beta de Linfotoxina/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Protocadherinas , Receptores de Interleucina-2/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Receptor Toll-Like 2/inmunología , Migración Transendotelial y Transepitelial/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/metabolismo
17.
J Clin Invest ; 130(5): 2602-2619, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32017712

RESUMEN

Lymph node stromal cells (LNSCs) regulate immunity through constructing lymphocyte niches. LNSC-produced laminin α5 (Lama5) regulates CD4+ T cells but the underlying mechanisms of its functions are poorly understood. Here we show that depleting Lama5 in LNSCs resulted in decreased Lama5 protein in the LN cortical ridge (CR) and around high endothelial venules (HEVs). Lama5 depletion affected LN structure with increased HEVs, upregulated chemokines, and cell adhesion molecules, and led to greater numbers of Tregs in the T cell zone. Mouse and human T cell transendothelial migration and T cell entry into LNs were suppressed by Lama5 through the receptors α6 integrin and α-dystroglycan. During immune responses and allograft transplantation, depleting Lama5 promoted antigen-specific CD4+ T cell entry into the CR through HEVs, suppressed T cell activation, and altered T cell differentiation to suppressive regulatory phenotypes. Enhanced allograft acceptance resulted from depleting Lama5 or blockade of T cell Lama5 receptors. Lama5 and Lama4/Lama5 ratios in allografts were associated with the rejection severity. Overall, our results demonstrated that stromal Lama5 regulated immune responses through altering LN structures and T cell behaviors. This study delineated a stromal Lama5-T cell receptor axis that can be targeted for immune tolerance modulation.


Asunto(s)
Laminina/inmunología , Ganglios Linfáticos/inmunología , Tolerancia al Trasplante/inmunología , Animales , Distroglicanos/metabolismo , Humanos , Integrina alfa6/metabolismo , Laminina/genética , Laminina/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/metabolismo , Vasos Linfáticos/citología , Vasos Linfáticos/inmunología , Vasos Linfáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células del Estroma/citología , Células del Estroma/inmunología , Células del Estroma/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Migración Transendotelial y Transepitelial/inmunología
18.
Methods Mol Biol ; 517: 145-67, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19378035

RESUMEN

Toll-like receptors (TLRs) are principal innate immune sensors critically involved in the recognition of evolutionary conserved microbial and viral structures called "pathogen-associated molecular patterns" (PAMPs). Although recognition patterns of many TLRs have been characterized, molecular mechanisms that initiate TLR signaling are poorly understood. Since posttranslational modifications of many receptor systems are important in initiating signaling, we studied whether tyrosine phosphorylation of TLR4, the principal sensor of Gram-negative bacterial lipopolysaccharide (LPS) plays a role in TLR4 signal-transducing functions. We found that LPS induced TLR4 tyrosine phosphorylation and mutations of tyrosine residues in the Toll-IL-1R signaling domain markedly suppressed TLR4-mediated activation of JNK and p38 MAP kinases and transcription factors NF-kappaB, RANTES, and IFN-beta. This chapter summarizes a combination of methodological approaches that can be used to demonstrate an indispensable role of TLR4 tyrosine phosphorylation in receptor signaling, including transient transfections, site-directed mutagenesis, immunoprecipitation and immunoblot analyses, and analyses of transcription factor activation in reporter assays.


Asunto(s)
Fosfotirosina/metabolismo , Receptor Toll-Like 4/metabolismo , Western Blotting , Línea Celular , Genes Reporteros/genética , Humanos , Mutagénesis Sitio-Dirigida , Fosforilación , Fosfotirosina/análisis , Fosfotirosina/genética , Plásmidos/genética , Transducción de Señal , Receptor Toll-Like 4/genética , Transgenes/genética
19.
Sci Immunol ; 4(33)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30877143

RESUMEN

Sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs) regulate migration of lymphocytes out of thymus to blood and lymph nodes (LNs) to efferent lymph, whereas their role in other tissue sites is not known. Here, we investigated the question of how these molecules regulate leukocyte migration from tissues through afferent lymphatics to draining LNs (dLNs). S1P, but not other chemokines, selectively enhanced human and murine CD4 T cell migration across lymphatic endothelial cells (LECs). T cell S1PR1 and S1PR4, and LEC S1PR2, were required for migration across LECs and into lymphatic vessels and dLNs. S1PR1 and S1PR4 differentially regulated T cell motility and vascular cell adhesion molecule-1 (VCAM-1) binding. S1PR2 regulated LEC layer structure, permeability, and expression of the junction molecules VE-cadherin, occludin, and zonulin-1 through the ERK pathway. S1PR2 facilitated T cell transcellular migration through VCAM-1 expression and recruitment of T cells to LEC migration sites. These results demonstrated distinct roles for S1PRs in comodulating T cell and LEC functions in migration and suggest previously unknown levels of regulation of leukocytes and endothelial cells during homeostasis and immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Movimiento Celular/inmunología , Células Endoteliales/inmunología , Vasos Linfáticos/inmunología , Receptores de Esfingosina-1-Fosfato/inmunología , Animales , Linfocitos T CD4-Positivos/fisiología , Línea Celular , Células Endoteliales/fisiología , Humanos , Lisofosfolípidos/inmunología , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Ratones Transgénicos , Esfingosina/análogos & derivados , Esfingosina/inmunología , Receptores de Esfingosina-1-Fosfato/genética , Proteínas de Uniones Estrechas/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología
20.
Nat Commun ; 10(1): 2927, 2019 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-31249309

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA