Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Nat Immunol ; 20(5): 593-601, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30886417

RESUMEN

Interferon-λ (IFN-λ) acts on mucosal epithelial cells and thereby confers direct antiviral protection. In contrast, the role of IFN-λ in adaptive immunity is far less clear. Here, we report that mice deficient in IFN-λ signaling exhibited impaired CD8+ T cell and antibody responses after infection with a live-attenuated influenza virus. Virus-induced release of IFN-λ triggered the synthesis of thymic stromal lymphopoietin (TSLP) by M cells in the upper airways that, in turn, stimulated migratory dendritic cells and boosted antigen-dependent germinal center reactions in draining lymph nodes. The IFN-λ-TSLP axis also boosted production of the immunoglobulins IgG1 and IgA after intranasal immunization with influenza virus subunit vaccines and improved survival of mice after challenge with virulent influenza viruses. IFN-λ did not influence the efficacy of vaccines applied by subcutaneous or intraperitoneal routes, indicating that IFN-λ plays a vital role in potentiating adaptive immune responses that initiate at mucosal surfaces.


Asunto(s)
Inmunidad Adaptativa/inmunología , Citocinas/inmunología , Inmunidad Mucosa/inmunología , Interleucinas/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/genética , Animales , Formación de Anticuerpos/efectos de los fármacos , Formación de Anticuerpos/inmunología , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/virología , Inmunidad Mucosa/efectos de los fármacos , Inmunidad Mucosa/genética , Inmunización/métodos , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Interleucinas/administración & dosificación , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Receptores de Interferón/metabolismo , Linfopoyetina del Estroma Tímico
2.
Immunity ; 53(5): 1015-1032.e8, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33207209

RESUMEN

Solitary intestinal lymphoid tissues such as cryptopatches (CPs) and isolated lymphoid follicles (ILFs) constitute steady-state activation hubs containing group 3 innate lymphoid cells (ILC3) that continuously produce interleukin (IL)-22. The outer surface of CPs and ILFs is demarcated by a poorly characterized population of CD11c+ cells. Using genome-wide single-cell transcriptional profiling of intestinal mononuclear phagocytes and multidimensional flow cytometry, we found that CP- and ILF-associated CD11c+ cells were a transcriptionally distinct subset of intestinal cDCs, which we term CIA-DCs. CIA-DCs required programming by CP- and ILF-resident CCR6+ ILC3 via lymphotoxin-ß receptor signaling in cDCs. CIA-DCs differentially expressed genes associated with immunoregulation and were the major cellular source of IL-22 binding protein (IL-22BP) at steady state. Mice lacking CIA-DC-derived IL-22BP exhibited diminished expression of epithelial lipid transporters, reduced lipid resorption, and changes in body fat homeostasis. Our findings provide insight into the design principles of an immunoregulatory checkpoint controlling nutrient absorption.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/inmunología , Receptores de Interleucina/biosíntesis , Animales , Biomarcadores , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inmunofenotipificación , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Metabolismo de los Lípidos , Ratones , Ratones Transgénicos , ARN Citoplasmático Pequeño/genética , Receptores de Interleucina/genética , Transducción de Señal
3.
Cell ; 157(2): 340-356, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725403

RESUMEN

Innate lymphoid cells (ILCs) are a recently recognized group of lymphocytes that have important functions in protecting epithelial barriers against infections and in maintaining organ homeostasis. ILCs have been categorized into three distinct groups, transcriptional circuitry and effector functions of which strikingly resemble the various T helper cell subsets. Here, we identify a common, Id2-expressing progenitor to all interleukin 7 receptor-expressing, "helper-like" ILC lineages, the CHILP. Interestingly, the CHILP differentiated into ILC2 and ILC3 lineages, but not into conventional natural killer (cNK) cells that have been considered an ILC1 subset. Instead, the CHILP gave rise to a peculiar NKp46(+) IL-7Rα(+) ILC lineage that required T-bet for specification and was distinct of cNK cells or other ILC lineages. Such ILC1s coproduced high levels of IFN-γ and TNF and protected against infections with the intracellular parasite Toxoplasma gondii. Our data significantly advance our understanding of ILC differentiation and presents evidence for a new ILC lineage that protects barrier surfaces against intracellular infections.


Asunto(s)
Diferenciación Celular , Linfocitos/citología , Linfocitos/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Factor de Transcripción GATA3/metabolismo , Inmunidad Innata , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-7/metabolismo , Células Madre/citología , Toxoplasma , Toxoplasmosis/inmunología
4.
EMBO J ; 39(13): e104159, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32627520

RESUMEN

γδ T cells with distinct properties develop in the embryonic and adult thymus and have been identified as critical players in a broad range of infections, antitumor surveillance, autoimmune diseases, and tissue homeostasis. Despite their potential value for immunotherapy, differentiation of γδ T cells in the thymus is incompletely understood. Here, we establish a high-resolution map of γδ T-cell differentiation from the fetal and adult thymus using single-cell RNA sequencing. We reveal novel sub-types of immature and mature γδ T cells and identify an unpolarized thymic population which is expanded in the blood and lymph nodes. Our detailed comparative analysis reveals remarkable similarities between the gene networks active during fetal and adult γδ T-cell differentiation. By performing a combined single-cell analysis of Sox13, Maf, and Rorc knockout mice, we demonstrate sequential activation of these factors during IL-17-producing γδ T-cell (γδT17) differentiation. These findings substantially expand our understanding of γδ T-cell ontogeny in fetal and adult life. Our experimental and computational strategy provides a blueprint for comparing immune cell differentiation across developmental stages.


Asunto(s)
Diferenciación Celular/inmunología , Feto/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Animales , Autoantígenos/genética , Autoantígenos/inmunología , Diferenciación Celular/genética , Ratones , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Linfocitos T/citología
5.
Transpl Int ; 37: 12263, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38550626

RESUMEN

ABO-incompatible (ABOi) living kidney transplantation (KTx) is an established procedure to address the demand for kidney transplants with outcomes comparable to ABO-compatible KTx. Desensitization involves the use of immunoadsorption (IA) to eliminate preformed antibodies against the allograft. This monocentric retrospective study compares single-use antigen-selective Glycosorb® ABO columns to reusable non-antigen-specific Immunosorba® immunoglobulin adsorption columns regarding postoperative infectious complications and outcome. It includes all 138 ABOi KTx performed at Freiburg Transplant Center from 2004-2020. We compare 81 patients desensitized using antigen-specific columns (sIA) to 57 patients who received IA using non-antigen-specific columns (nsIA). We describe distribution of infections, mortality and allograft survival in both groups and use Cox proportional hazards regression to test for the association of IA type with severe infections. Desensitization with nsIA tripled the risk of severe postoperative infections (adjusted HR 3.08, 95% CI: 1.3-8.1) compared to sIA. nsIA was associated with significantly more recurring (21.4% vs. 6.2%) and severe infections (28.6% vs. 8.6%), mostly in the form of urosepsis. A significantly higher proportion of patients with sIA suffered from allograft rejection (29.6% vs. 14.0%). However, allograft survival was comparable. nsIA is associated with a two-fold risk of developing a severe postoperative infection after ABOi KTx.


Asunto(s)
Trasplante de Riñón , Humanos , Trasplante de Riñón/efectos adversos , Trasplante de Riñón/métodos , Estudios Retrospectivos , Sistema del Grupo Sanguíneo ABO , Incompatibilidad de Grupos Sanguíneos , Factores de Riesgo , Rechazo de Injerto , Supervivencia de Injerto , Donadores Vivos
6.
J Immunol ; 208(7): 1585-1594, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35296538

RESUMEN

Innate lymphoid cells (ILCs) that express NK cell receptors (NCRs) and the transcription factor T-bet populate nonlymphoid tissues and are crucial in immune responses against viral infections and malignancies. Recent studies highlighted the heterogeneity of this ILC population and extended their functional spectrum to include important roles in tissue homeostasis and autoimmunity. In this article, we provide detailed profiling of NCR+T-bet+ ILC populations in the murine kidney, identifying conventional NK (cNK) cells and type 1 ILCs (ILC1s) as the two major subsets. Induction of renal inflammation in a mouse model of glomerulonephritis did not substantially influence abundance or phenotype of cNK cells or ILC1s in the kidney. For functional analyses in this model, widely used depletion strategies for total NCR+ ILCs (anti-NK1.1 Ab application) and cNK cells (anti-asialoGM1 serum application) were unreliable tools, because they were accompanied by significant off-target depletion of kidney NKT cells and CD8+ T cells, respectively. However, neither depletion of cNK cells and ILC1s in NKT cell-deficient mice nor specific genetic deletion of cNK cells in Ncr1 Cre/wt × Eomes fl/fl mice altered the clinical course of experimental glomerulonephritis. In summary, we show in this article that cNK cells and ILC1s are dispensable for initiation and progression of immune-mediated glomerular disease and advise caution in the use of standard Ab depletion methods to study NCR+ ILC function in mouse models.


Asunto(s)
Glomerulonefritis , Inmunidad Innata , Animales , Linfocitos T CD8-positivos , Riñón , Células Asesinas Naturales , Ratones
7.
Eur J Immunol ; 52(9): 1406-1418, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35339113

RESUMEN

The intestinal tract is the largest immunological organ in the body and has a central function of regulating local immune responses, as the intestinal epithelial barrier is a location where the immune system interacts with the gut microbiome including bacteria, fungi, and viruses. Impaired immunity in the intestinal tract can lead to immunopathology, which manifests in different diseases such as inflammatory bowel disease or intestinal graft-versus-host disease. A disturbed communication between epithelial cells, immune cells, and microbiome will shape pathogenic immune responses to antigens, which need to be counterbalanced by tolerogenic mechanisms and repair mechanisms. Here, we review how impaired intestinal immune function leads to immunopathology with a specific focus on innate immune cells, the role of the microbiome, and the resulting clinical manifestations including intestinal graft-versus-host disease, inflammatory bowel disease, and enteropathy in primary immunodeficiency.


Asunto(s)
Microbioma Gastrointestinal , Enfermedad Injerto contra Huésped , Enfermedades Inflamatorias del Intestino , Microbiota , Células Epiteliales/patología , Enfermedad Injerto contra Huésped/patología , Humanos , Inmunidad Innata , Mucosa Intestinal
8.
Immunity ; 41(2): 230-43, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25148024

RESUMEN

CD8αα(+) intraepithelial lymphocytes (IELs) are instrumental in maintaining the epithelial barrier in the intestine. Similar to natural killer cells and other innate lymphoid cells, CD8αα(+) IELs constitutively express the T-box transcription factor T-bet. However, the precise role of T-bet for the differentiation or function of IELs is unknown. Here we show that mice genetically deficient for T-bet lacked both TCRαß(+) and TCRγδ(+) CD8αα(+) IELs and thus are more susceptible to chemically induced colitis. Although T-bet was induced in thymic IEL precursors (IELPs) as a result of agonist selection and interleukin-15 (IL-15) receptor signaling, it was dispensable for the generation of IELPs. Subsequently, T-bet was required for the IL-15-dependent activation, differentiation, and expansion of IELPs in the periphery. Our study reveals a function of T-bet as a central transcriptional regulator linking agonist selection and IL-15 signaling with the emergence of CD8αα(+) IELs.


Asunto(s)
Antígenos CD8/biosíntesis , Interleucina-15/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Proteínas de Dominio T Box/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Células Epiteliales/inmunología , Interleucina-15/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Intestinos/citología , Intestinos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-15/inmunología , Transducción de Señal/inmunología , Proteínas de Dominio T Box/biosíntesis
9.
BMC Nephrol ; 24(1): 99, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-37061677

RESUMEN

BACKGROUND: Despite vaccination coronavirus disease 2019 (COVID-19)-associated mortality caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains high in kidney transplant recipients. Nirmatrelvir is a protease inhibitor with activity against SARS-CoV-2. Nirmatrelvir reduces the risk for mortality and hospitalization, which is approved for treating adults at risk for severe disease. Nirmatrelvir is metabolized by the cytochrome P-450 (CYP) 3A4 isozyme CYP3A4 and is therefore co-administered with the irreversible CYP3A4 inhibitor ritonavir, which results in a drug interaction with tacrolimus. A limited number of patients with nirmatrelvir/ritonavir and tacrolimus therapy after kidney transplantation have been reported to date. It has been reported that tacrolimus was paused during the five-day nirmatrelvir/ritonavir therapy and subtherapeutic tacrolimus levels were observed after finishing nirmatrelvir/ritonavir in two patients. Therefore, optimization of tacrolimus dosing is urgently needed in transplant recipients receiving nirmatrelvir/ritonavir treatment. CASE PRESENTATION: Here, we present our first-hand experience with four patients receiving tacrolimus therapy following kidney transplantation and nirmatrelvir/ritonavir therapy due to COVID-19. Tacrolimus was paused during nirmatrelvir/ritonavir therapy in all patients, which resulted in stable therapeutic tacrolimus levels. Tacrolimus was continued directly after finishing nirmatrelvir/ritonavir to avoid subtherapeutic levels in the first patient treated. This patient received his usual tacrolimus maintenance dose, which resulted in toxic levels. Based on this observation, tacrolimus therapy was continued 24 h after finishing nirmatrelvir/ritonavir treatment at a reduced dose in the subsequent patients. In these patients, therapeutic to supratherapeutic tacrolimus levels were observed despite the therapeutic break and dose reduction. DISCUSSION AND CONCLUSIONS: Based on altered CYP3A4 metabolism, tacrolimus levels have to be closely monitored after treatment with nirmatrelvir/ritonavir. Our study suggests that tacrolimus treatment should be paused during nirmatrelvir/ritonavir medication and be continued 24 h after completing nirmatrelvir/ritonavir therapy at a reduced dose and under close monitoring. Based on the limited number of patients in this study, results must be interpreted with caution.


Asunto(s)
COVID-19 , Trasplante de Riñón , Adulto , Humanos , Citocromo P-450 CYP3A , SARS-CoV-2 , Ritonavir/uso terapéutico , Tacrolimus/uso terapéutico , Receptores de Trasplantes , Tratamiento Farmacológico de COVID-19 , Antivirales/uso terapéutico
10.
J Hepatol ; 77(4): 978-990, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35636577

RESUMEN

BACKGROUND & AIMS: In immunosuppressed patients, persistent HEV infection is common and may lead to cirrhosis and liver failure. HEV clearance depends on an effective virus-specific CD8+ T-cell response; however, the knowledge gap around HEV-specific CD8+ T-cell epitopes has hindered analysis of the mechanisms of T-cell failure in persistent infection. METHODS: We comprehensively studied HEV-specific CD8+ T-cell responses in 46 patients with self-limiting (n = 34) or chronic HEV infection (n = 12), by epitope-specific expansion, functional testing, ex vivo peptide HLA class I tetramer multi-parametric staining, and viral sequence analysis. RESULTS: We identified 25 HEV-specific CD8+ T-cell epitopes restricted by 9 different HLA class I alleles. In self-limiting HEV infection, HEV-specific CD8+ T cells were vigorous, contracted after resolution of infection, and formed functional memory responses. In contrast, in chronic infection, the HEV-specific CD8+ T-cell response was diminished, declined over time, and displayed phenotypic features of exhaustion. However, improved proliferation of HEV-specific CD8+ T cells, increased interferon-γ production and evolution of a memory-like phenotype were observed upon reduction of immunosuppression and/or ribavirin treatment and were associated with viral clearance. In 1 patient, mutational viral escape in a targeted CD8+ T-cell epitope contributed to CD8+ T-cell failure. CONCLUSION: Chronic HEV infection is associated with HEV-specific CD8+ T-cell exhaustion, indicating that T-cell exhaustion driven by persisting antigen recognition also occurs in severely immunosuppressed hosts. Functional reinvigoration of virus-specific T cells is at least partially possible when antigen is cleared. In a minority of patients, viral escape also contributes to HEV-specific CD8+ T-cell failure and thus needs to be considered in personalized immunotherapeutic approaches. LAY SUMMARY: Hepatitis E virus (HEV) infection is usually cleared spontaneously (without treatment) in patients with fully functioning immune systems. In immunosuppressed patients, chronic HEV infection is common and can progress rapidly to cirrhosis and liver failure. Herein, we identified the presence of HEV-specific CD8+ T cells (a specific type of immune cell that can target HEV) in immunosuppressed patients, but we show that these cells do not function properly. This dysfunction appears to play a role in the development of chronic HEV infection in vulnerable patients.


Asunto(s)
Virus de la Hepatitis E , Hepatitis E , Fallo Hepático , Linfocitos T CD8-positivos , Epítopos de Linfocito T , Humanos , Interferón gamma , Cirrosis Hepática , Ribavirina
11.
PLoS Pathog ; 16(9): e1008870, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32991634

RESUMEN

The two T-box transcription factors T-bet and Eomesodermin (Eomes) are important regulators of cytotoxic lymphocytes (CTLs), such as activated CD8 T cells, which are essential in the fight against intracellular pathogens and tumors. Both transcription factors share a great degree of homology based on sequence analysis and as a result exert partial functional redundancy during viral infection. However, the actual degree of redundancy between T-bet and Eomes remains a matter of debate and is further confounded by their distinct spatiotemporal expression pattern in activated CD8 T cells. To directly investigate the functional overlap of these transcription factors, we generated a new mouse model in which Eomes expression is under the transcriptional control of the endogenous Tbx21 (encoding for T-bet) locus. Applying this model, we demonstrate that the induction of Eomes in lieu of T-bet cannot rescue T-bet deficiency in CD8 T cells during acute lymphocytic choriomeningitis virus (LCMV) infection. We found that the expression of Eomes instead of T-bet was not sufficient for early cell expansion or effector cell differentiation. Finally, we show that imposed expression of Eomes after acute viral infection promotes some features of exhaustion but must act in concert with other factors during chronic viral infection to establish all hallmarks of exhaustion. In summary, our results clearly underline the importance of T-bet in guiding canonical CTL development during acute viral infections.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/fisiología , Proteínas de Dominio T Box/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Proteínas Fetales/metabolismo , Regulación de la Expresión Génica/fisiología , Interferón gamma/metabolismo , Ratones Transgénicos
12.
Clin Transplant ; 36(7): e14663, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35368114

RESUMEN

BACKGROUND: BK polyoma virus (BKPyV) associated nephropathy (BKPyVAN) is a major cause of kidney graft loss in renal transplant patients. Interferons (IFNs) are an important innate immune response against viral infections and genetic polymorphisms of the IFN-pathways can affect susceptibility and mortality during viral infection. Here, we investigated whether the dinucleotide polymorphism rs368234815 (ΔG/TT) in the IFNL4 gene contributed to BKPyV reactivation or BKPyVAN after living-donor kidney transplantation. METHODS: This retrospective case-control study determines the prevalence of IFNL4 variants in a Caucasian population of living-donor kidney transplant recipients and donors and explores its association with BKPyV infection and BKPyVAN development. We included 28 recipients with BKPyV reactivation, 10 of which developed BKPyVAN and 30 BKPyV negative controls. Targeted sequencing of the IFNL4 gene from both recipients and their respective donors was performed. RESULTS: We found IFNL4 rs368234815 ΔG allele frequencies of 41.7% in BKPyV negative and 39.3% in BKPyV positive recipients (P = .85), and 41.7% and 40.4% (P>.99) in their respective donors. IFNL4 rs368234815 ΔG allele frequencies in BKPyVAN developing recipients and their respective donors were 50% and 43.7% (P = .60 and P>.99). CONCLUSIONS: Our results indicate that the IFNL4 rs368234815 ΔG allele is not associated with BKPyV reactivation, nor the manifestation of BKPyVAN.


Asunto(s)
Virus BK , Trasplante de Riñón , Infecciones por Polyomavirus , Infecciones Tumorales por Virus , Estudios de Casos y Controles , Humanos , Interleucinas , Trasplante de Riñón/efectos adversos , Trasplante de Riñón/métodos , Donadores Vivos , Polimorfismo Genético , Infecciones por Polyomavirus/epidemiología , Infecciones por Polyomavirus/genética , Estudios Retrospectivos , Receptores de Trasplantes , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/genética
13.
BMC Infect Dis ; 22(1): 734, 2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36104663

RESUMEN

BACKGROUND: Effective SARS-CoV-2 vaccination in patients receiving treatment with B-cell depleting agents is challenging. Information on vaccination responses in these patients are a valuable tool to develop efficient vaccination regimens. METHODS: In this single-center retrospective observational study, we report the humoral and cellular response in 34 patients receiving anti-CD20 antibody treatment for renal immune disease. RESULTS: After base immunization with SARS-CoV-2-vaccines, 92.0% developed a cellular, 32.4% a humoral response. Humoral immunity correlated with B-cell counts and the timespan between anti-CD20 antibody treatment and vaccination. All patients with > 21/µl B-cells, or > 197 days after treatment showed humoral response. CONCLUSIONS: Adequate timing of SARS-CoV-2-vaccinations after anti-CD20 antibody treatment and CD19 measurements are crucial to generate immunity. Awaiting partial B-cell recovery by postponing regularly scheduled anti-CD20 treatment should be considered in patients with stable immune disease. TRIAL REGISTRATION: This study has been retrospectively registered in the German Clinical Trials Register (DRKS00027049) on 29/10/2021.


Asunto(s)
Enfermedades Autoinmunes , COVID-19 , Vacunas Virales , Anticuerpos Antivirales/uso terapéutico , Vacunas contra la COVID-19 , Humanos , SARS-CoV-2
14.
Infect Immun ; 89(11): e0080020, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34424753

RESUMEN

Innate lymphoid cells (ILCs) comprise five distinct subsets. ILCs are found at mucosal barriers and may fight invading pathogens. Chlamydia is an intracellular bacterium that infects the mucosa of the genital tract and can cause severe tissue damage. Here, we used a mouse infection model with Chlamydia muridarum to measure the reaction of genital tract ILCs to the infection. Tissue-resident natural killer (NK) cells were the largest group in the uninfected female genital tract, and their number did not substantially change. Conventional NK cells were present in the greatest numbers during acute infection, while ILC1s continuously increased to high numbers. ILC2 and ILC3s were found at lower numbers that oscillated by a factor of 2 to 4. The majority of ILC3s transdifferentiated into ILC1s. NK cells and ILC1s produced gamma interferon (IFN-γ) and, rarely, tumor necrosis factor (TNF), but only early in the infection. Lack of B and T cells increased ILC numbers, while the loss of myeloid cells decreased them. ILCs accumulated to a high density in the oviduct, a main site of tissue destruction. ILC subsets are part of the inflammatory and immune reaction during infection with C. muridarum and may contribute to tissue damage during chlamydial infection.


Asunto(s)
Infecciones por Chlamydia/inmunología , Genitales Femeninos/inmunología , Linfocitos/inmunología , Animales , Femenino , Inmunidad Innata , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL
15.
BMC Nephrol ; 21(1): 486, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33198670

RESUMEN

BACKGROUND: Critically ill coronavirus disease 2019 (COVID-19) patients have a high risk of acute kidney injury (AKI) that requires renal replacement therapy (RRT). A state of hypercoagulability reduces circuit life spans. To maintain circuit patency and therapeutic efficiency, an optimized anticoagulation strategy is needed. This study investigates whether alternative anticoagulation strategies for RRT during COVID-19 are superior to administration of unfractionated heparin (UFH). METHODS: Retrospective cohort study on 71 critically ill COVID-19 patients (≥18 years), admitted to intensive care units at a tertiary health care facility in the southwestern part of Germany between February 26 and May 21, 2020. We collected data on the disease course, AKI, RRT, and thromboembolic events. Four different anticoagulatory regimens were administered. Anticoagulation during continuous veno-venous hemodialysis (CVVHD) was performed with UFH or citrate. Anticoagulation during sustained low-efficiency daily dialysis (SLEDD) was performed with UFH, argatroban, or low molecular weight heparin (LMWH). Primary outcome is the effect of the anticoagulation regimen on mean treatment times of RRT. RESULTS: In patients receiving CVVHD, mean treatment time in the UFH group was 21.3 h (SEM: ±5.6 h), in the citrate group 45.6 h (SEM: ±2.7 h). Citrate anticoagulation significantly prolonged treatment times by 24.4 h (P = .001). In patients receiving SLEDD, mean treatment time with UFH was 8.1 h (SEM: ±1.3 h), with argatroban 8.0 h (SEM: ±0.9 h), and with LMWH 11.8 h (SEM: ±0.5 h). LMWH significantly prolonged treatment times by 3.7 h (P = .008) and 3.8 h (P = .002), respectively. CONCLUSIONS: UFH fails to prevent early clotting events in the dialysis circuit during COVID-19. For patients, who do not require effective systemic anticoagulation, regional citrate dialysis is the most effective strategy. For patients, who require effective systemic anticoagulation, the usage of LMWH results in the longest circuit life spans. The proposed anticoagulatory strategies are safe, can easily be monitored, and allow an individualized treatment.


Asunto(s)
Lesión Renal Aguda/terapia , Anticoagulantes/administración & dosificación , Betacoronavirus , Infecciones por Coronavirus/epidemiología , Neumonía Viral/epidemiología , Terapia de Reemplazo Renal/métodos , Lesión Renal Aguda/sangre , Lesión Renal Aguda/epidemiología , Adulto , Anciano , Arginina/análogos & derivados , Coagulación Sanguínea , COVID-19 , Ácido Cítrico/administración & dosificación , Comorbilidad , Infecciones por Coronavirus/sangre , Cuidados Críticos , Enfermedad Crítica , Falla de Equipo , Femenino , Alemania/epidemiología , Heparina/administración & dosificación , Heparina de Bajo-Peso-Molecular/administración & dosificación , Humanos , Masculino , Persona de Mediana Edad , Pandemias , Ácidos Pipecólicos/administración & dosificación , Neumonía Viral/sangre , Terapia de Reemplazo Renal/instrumentación , Estudios Retrospectivos , SARS-CoV-2 , Sulfonamidas , Centros de Atención Terciaria
16.
Nature ; 494(7436): 261-5, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23334414

RESUMEN

At mucosal surfaces, the immune system should not initiate inflammatory immune responses to the plethora of antigens constantly present in the environment, but should remain poised to unleash a potent assault on intestinal pathogens. The transcriptional programs and regulatory factors required for immune cells to switch from homeostatic (often tissue-protective) function to potent antimicrobial immunity are poorly defined. Mucosal retinoic-acid-receptor-related orphan receptor-γt-positive (RORγt(+)) innate lymphoid cells (ILCs) are emerging as an important innate lymphocyte population required for immunity to intestinal infections. Various subsets of RORγt(+) ILCs have been described but the transcriptional programs controlling their specification and fate remain largely unknown. Here we provide evidence that the transcription factor T-bet determines the fate of a distinct lineage of CCR6(-)RORγt(+) ILCs. Postnatally emerging CCR6(-)RORγt(+) ILCs upregulated T-bet and this was controlled by cues from the commensal microbiota and interleukin-23 (IL-23). In contrast, CCR6(+)RORγt(+) ILCs, which arise earlier during ontogeny, did not express T-bet. T-bet instructed the expression of T-bet target genes such as interferon-γ (IFN-γ) and of the natural cytotoxicity receptor NKp46. Mice genetically lacking T-bet showed normal development of CCR6(-)RORγt(+) ILCs, but they could not differentiate into NKp46-expressing RORγt(+) ILCs (that is, IL-22-producing natural killer (NK-22) cells) and failed to produce IFN-γ. The production of IFN-γ by T-bet-expressing CCR6(-)RORγt(+) ILCs was essential for the release of mucus-forming glycoproteins required to protect the epithelial barrier during Salmonella enterica infection. Salmonella infection also causes severe enterocolitis that is at least partly driven by IFN-γ. Mice deficient for T-bet or depleted of ILCs developed only mild enterocolitis. Thus, graded expression of T-bet in CCR6(-)RORγt(+) ILCs facilitates the differentiation of IFN-γ-producing CCR6(-)RORγt(+) ILCs required to protect the epithelial barrier against Salmonella infections. Co-expression of T-bet and RORγt, which is also found in subsets of IL-17-producing T-helper (T(H)17) cells, may be an evolutionarily conserved transcriptional program that originally developed as part of the innate defence against infections but that also confers an increased risk of immune-mediated pathology.


Asunto(s)
Linaje de la Célula , Inmunidad Innata/inmunología , Linfocitos/citología , Linfocitos/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores CCR6/deficiencia , Proteínas de Dominio T Box/metabolismo , Animales , Antígenos Ly/genética , Diferenciación Celular , Células Cultivadas , Enterocolitis/inmunología , Enterocolitis/metabolismo , Enterocolitis/patología , Epitelio/inmunología , Epitelio/metabolismo , Epitelio/microbiología , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-23/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Moco/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Receptores CCR6/metabolismo , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad
17.
Genesis ; 55(8)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28646547

RESUMEN

The T-box transcription factor Eomes (also known as Tbr2) shows short-lived expression in various localized domains of the embryo, including epiblast cells during gastrulation and intermediate progenitor cells in the cerebral cortex. In these tissues Eomes fulfills crucial roles for lineage specification of progenitors. To directly observe Eomes-dependent cell lineages in the living embryo, we generated a novel dual-fluorescence reporter allele that expresses a membrane-bound tdTomato protein for investigation of cell morphology and a nuclear GFP for cell tracing. This allele recapitulates endogenous EOMES protein expression and is suitable for live imaging. We found that the allele can also be used as a short-to-medium-term lineage tracer, as GFP persists in cells longer than EOMES protein and marks Eomes-dependent lineages with a timeframe of days to weeks depending on the proliferation rate. In summary, we present a novel genetic tool for investigation of Eomes-dependent cell types by live imaging and lineage tracing.


Asunto(s)
Linaje de la Célula , Rastreo Celular/métodos , Genes Reporteros , Ingeniería Genética/métodos , Imagen Óptica/métodos , Proteínas de Dominio T Box/genética , Animales , Femenino , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Proteínas de Dominio T Box/metabolismo
18.
J Clin Immunol ; 37(8): 759-769, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28936778

RESUMEN

PURPOSE: Over a third of patients with common variable immunodeficiency (CVID) suffer from secondary complications like inflammatory organ disease, autoimmune manifestations, or lymphoproliferation contributing to increased morbidity and mortality in affected patients. Innate lymphoid cells (ILCs) have emerging roles in setting the milieu for physiological, but also pathological, immune responses and inflammation. We therefore sought to correlate the recently identified disturbed homeostasis of ILCs with alterations of the adaptive immune system in complex CVID patients (CVIDc). METHODS: We quantified peripheral blood ILC and T helper cell subsets of 58 CVID patients by flow cytometry and compared the results to the clinical and immunological phenotype. RESULTS: Total ILCs were significantly reduced in peripheral blood of CVIDc patients compared to healthy individuals, but not to CVID patients who suffered only from infections (CVIDio). This reduction was mainly due to a decrease in ILC2s, while ILC3s were relatively increased in CVIDc compared to CVIDio patients. This alteration in ILC phenotype was more prominent in patients with an expansion of CD21low B cells, but we could not detect an association of the altered ILC phenotype with a TH1-shift among circulating CD4 T cells, which was also prominent in CVIDc patients. CONCLUSION: We confirm a relative shift in ILCs of CVIDc patients towards ILC3s which was associated with the expansion of CD21low B cells, but not overtly with the relative expansion of TH1-like T cells. Given the relative abundance of TH1-like T cells compared to ILCs, these probably represent a more prominent source of the observed IFNγ-signature in CVIDc patients.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Inmunodeficiencia Variable Común/inmunología , Inmunidad Innata , Linfocitos/inmunología , Trastornos Linfoproliferativos/inmunología , Inmunidad Adaptativa , Adulto , Anciano , Enfermedades Autoinmunes/etiología , Circulación Sanguínea , Inmunodeficiencia Variable Común/complicaciones , Citocinas/metabolismo , Femenino , Homeostasis , Humanos , Tolerancia Inmunológica , Inflamación , Trastornos Linfoproliferativos/etiología , Masculino , Persona de Mediana Edad , Fenotipo , Células Th2/inmunología , Adulto Joven
19.
Int Immunol ; 26(3): 119-28, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24585669

RESUMEN

Innate lymphoid cells (ILCs) are a heterogeneous group of lymphocytes, which play an important role in tissue homeostasis at epithelial surfaces. They are scarce in spleen and lymph nodes, but substantial numbers can be found in the intestinal mucosa even at steady state. There, they represent the first line of defence against invading pathogens and contribute to lymphorganogenesis, tissue repair and, when inappropriately activated, immune pathology. Lineage-specific development, function and maintenance of these cells depend on a restricted set of transcription factors that partially emerged as a result of diversification and selection during vertebrate evolution. The differential expression of transcription factors regulates unique developmental programs, which endow the different ILC subsets with specific effector functions. Despite this division of labour, ILCs are considered to share a common origin, as they all are progeny of the common lymphoid progenitor, rely on the common γ-chain (γc) used by various cytokine receptors and show a developmental requirement for the transcriptional regulator Id2 (inhibitor of DNA binding 2). Here, we review the transcriptional programs required for the development and function of ILCs and give an overview of the evolution of transcription factors and cytokines expressed by ILCs.


Asunto(s)
Mucosa Intestinal/inmunología , Linfocitos/inmunología , Factores de Transcripción/inmunología , Animales , Evolución Biológica , Diferenciación Celular , Linaje de la Célula , Citocinas/metabolismo , Regulación del Desarrollo de la Expresión Génica/inmunología , Homeostasis , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/genética , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Activación de Linfocitos/genética , Factores de Transcripción/genética
20.
J Immunol ; 190(9): 4848-60, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23536635

RESUMEN

Modification of allogeneic dendritic cells (DCs) through drug treatment results in DCs with in vitro hallmarks of tolerogenicity. Despite these observations, using murine MHC-mismatched skin and heart transplant models, donor-derived drug-modified DCs not only failed to induce tolerance but also accelerated graft rejection. The latter was inhibited by injecting the recipient with anti-CD8 Ab, which removed both CD8(+) T cells and CD8(+) DCs. The discrepancy between in vitro and in vivo data could be explained, partly, by the presentation of drug-modified donor DC MHC alloantigens by recipient APCs and activation of recipient T cells with indirect allospecificity, leading to the induction of alloantibodies. Furthermore, allogeneic MHC molecules expressed by drug-treated DCs were rapidly processed and presented in peptide form by recipient APCs in vivo within hours of DC injection. Using TCR-transgenic T cells, Ag presentation of injected OVA-pulsed DCs was detectable for ≤ 3 d, whereas indirect presentation of MHC alloantigen by recipient APCs led to activation of T cells within 14 h and was partially inhibited by reducing the numbers of CD8(+) DCs in vivo. In support of this observation when mice lacking CD8(+) DCs were pretreated with drug-modified DCs prior to transplantation, skin graft rejection kinetics were similar to those in non-DC-treated controls. Of interest, when the same mice were treated with anti-CD40L blockade plus drug-modified DCs, skin graft survival was prolonged, suggesting endogenous DCs were responsible for T cell priming. Altogether, these findings highlight the risks and limitations of negative vaccination using alloantigen-bearing "tolerogenic" DCs.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células Dendríticas/inmunología , Tolerancia Inmunológica/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Isoantígenos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/inmunología , Trasplante de Piel/inmunología , Donantes de Tejidos , Trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA