Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Immunol ; 9(99): eadn2362, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39241054

RESUMEN

Mucosal-associated invariant T (MAIT) cells are unconventional T cells that recognize microbial riboflavin pathway metabolites presented by evolutionarily conserved MR1 molecules. We explored the human MAIT cell compartment across organ donor-matched blood, barrier, and lymphoid tissues. MAIT cell population size was donor dependent with distinct tissue compartmentalization patterns and adaptations: Intestinal CD103+ resident MAIT cells presented an immunoregulatory CD39highCD27low profile, whereas MAIT cells expressing NCAM1/CD56 dominated in the liver and exhibited enhanced effector capacity with elevated response magnitude and polyfunctionality. Both intestinal CD39high and hepatic CD56+ adaptations accumulated with donor age. CD56+ MAIT cells displayed limited T cell receptor-repertoire breadth, elevated MR1 binding, and a transcriptional profile skewed toward innate activation pathways. Furthermore, CD56 was dynamically up-regulated to a persistent steady-state equilibrium after exposure to antigen or IL-7. In summary, we demonstrate functional heterogeneity and tissue site adaptation in resident MAIT cells across human barrier tissues with distinct regulatory and effector signatures.


Asunto(s)
Células T Invariantes Asociadas a Mucosa , Humanos , Células T Invariantes Asociadas a Mucosa/inmunología , Adulto , Masculino , Femenino , Persona de Mediana Edad , Antígenos CD/inmunología , Hígado/inmunología
2.
Nat Immunol ; 25(8): 1445-1459, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38956379

RESUMEN

The functional diversity of natural killer (NK) cell repertoires stems from differentiation, homeostatic, receptor-ligand interactions and adaptive-like responses to viral infections. In the present study, we generated a single-cell transcriptional reference map of healthy human blood- and tissue-derived NK cells, with temporal resolution and fate-specific expression of gene-regulatory networks defining NK cell differentiation. Transfer learning facilitated incorporation of tumor-infiltrating NK cell transcriptomes (39 datasets, 7 solid tumors, 427 patients) into the reference map to analyze tumor microenvironment (TME)-induced perturbations. Of the six functionally distinct NK cell states identified, a dysfunctional stressed CD56bright state susceptible to TME-induced immunosuppression and a cytotoxic TME-resistant effector CD56dim state were commonly enriched across tumor types, the ratio of which was predictive of patient outcome in malignant melanoma and osteosarcoma. This resource may inform the design of new NK cell therapies and can be extended through transfer learning to interrogate new datasets from experimental perturbations or disease conditions.


Asunto(s)
Células Asesinas Naturales , Linfocitos Infiltrantes de Tumor , Neoplasias , Transcriptoma , Microambiente Tumoral , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Microambiente Tumoral/inmunología , Neoplasias/inmunología , Neoplasias/genética , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Perfilación de la Expresión Génica , Análisis de la Célula Individual , Redes Reguladoras de Genes , Antígeno CD56/metabolismo , Regulación Neoplásica de la Expresión Génica , Diferenciación Celular
3.
Oncoimmunology ; 12(1): 2233402, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37448786

RESUMEN

Lung cancer is a leading cause of cancer-related death worldwide. Despite recent advances in tissue immunology, little is known about the spatial distribution of tissue-resident lymphocyte subsets in lung tumors. Using high-parameter flow cytometry, we identified an accumulation of tissue-resident lymphocytes including tissue-resident NK (trNK) cells and CD8+ tissue-resident memory T (TRM) cells toward the center of human non-small cell lung carcinomas (NSCLC). Chemokine receptor expression patterns indicated different modes of tumor-infiltration and/or residency between trNK cells and CD8+ TRM cells. In contrast to CD8+ TRM cells, trNK cells and ILCs generally expressed low levels of immune checkpoint receptors independent of location in the tumor. Additionally, granzyme expression in trNK cells and CD8+ TRM cells was highest in the tumor center, and intratumoral CD49a+CD16- NK cells were functional and responded stronger to target cell stimulation than their CD49a- counterparts, indicating functional relevance of trNK cells in lung tumors. In summary, the present spatial mapping of lymphocyte subsets in human NSCLC provides novel insights into the composition and functionality of tissue-resident immune cells, suggesting a role for trNK cells and CD8+ TRM cells in lung tumors and their potential relevance for future therapeutic approaches.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Linfocitos T CD8-positivos , Inmunidad Innata , Integrina alfa1/metabolismo , Células Asesinas Naturales/metabolismo
4.
Front Immunol ; 13: 834862, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35371005

RESUMEN

Respiratory viral infections with SARS-CoV-2 and influenza viruses commonly induce a strong infiltration of immune cells into the human lung, with potential detrimental effects on the integrity of the lung tissue. Despite comprising the largest fractions of circulating lymphocytes in the lung, rather little is known about how peripheral blood natural killer (NK) cell and T cell subsets are equipped for lung-homing in COVID-19 and influenza. Here, we provide a detailed comparative analysis of NK cells and T cells in patients infected with SARS-CoV-2 or influenza virus, focusing on the protein and gene expression of chemokine receptors known to be involved in recruitment to the lung. For this, we used 28-colour flow cytometry as well as re-analysis of a publicly available single-cell RNA-seq dataset from bronchoalveolar lavage (BAL) fluid. Frequencies of NK cells and T cells expressing CXCR3, CXCR6, and CCR5 were altered in peripheral blood of COVID-19 and influenza patients, in line with increased transcript expression of CXCR3, CXCR6, and CCR5 and their respective ligands in BAL fluid. NK cells and T cells expressing lung-homing receptors displayed stronger phenotypic signs of activation compared to cells lacking lung-homing receptors, and activation was overall stronger in influenza compared to COVID-19. Together, our results indicate a role for CXCR3+, CXCR6+, and/or CCR5+ NK cells and T cells that potentially migrate to the lungs in moderate COVID-19 and influenza patients, identifying common targets for future therapeutic interventions in respiratory viral infections.


Asunto(s)
COVID-19 , Gripe Humana , Expresión Génica , Humanos , Gripe Humana/metabolismo , Células Asesinas Naturales , Pulmón , SARS-CoV-2 , Subgrupos de Linfocitos T
5.
Cell Rep Med ; 3(2): 100508, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35243416

RESUMEN

Few approaches have been made toward exploring autologous NK cells in settings of cancer immunotherapy. Here, we demonstrate the feasibility of infusing multiple doses of ex vivo activated and expanded autologous NK cells in patients with multiple myeloma (MM) post-autologous stem cell transplantation. Infused NK cells were detected in circulation up to 4 weeks after the last infusion. Elevations in plasma granzyme B levels were observed following each consecutive NK cell infusion. Moreover, increased granzyme B levels were detected in bone marrow 4 weeks after the last infusion. All measurable patients had objective, detectable responses after NK cell infusions in terms of reduction in M-component and/or minimal residual disease. The present study demonstrates that autologous NK cell-based immunotherapy is feasible in a setting of MM consolidation therapy. It opens up the possibility for usage of autologous NK cells in clinical settings where patients are not readily eligible for allogeneic NK cell-based immunotherapies.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mieloma Múltiple , Quimioterapia de Consolidación , Granzimas , Humanos , Células Asesinas Naturales , Mieloma Múltiple/terapia , Trasplante de Células Madre , Trasplante Autólogo
6.
Front Immunol ; 12: 752104, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867984

RESUMEN

Innate lymphoid cells (ILCs) contribute to immune defense, yet it is poorly understood how ILCs develop and are strategically positioned in the lung. This applies especially to human ILCs due to the difficulty of studying them in vivo. Here we investigated the ontogeny and migration of human ILCs in vivo with a humanized mouse model ("MISTRG") expressing human cytokines. In addition to known tissue-resident ILC subsets, we discovered CD5-expressing ILCs that predominantly resided within the lung vasculature and in the circulation. CD5+ ILCs contained IFNγ-producing mature ILC1s as well as immature ILCs that produced ILC effector cytokines under polarizing conditions in vitro. CD5+ ILCs had a distinct ontogeny compared to conventional CD5- ILCs because they first appeared in the thymus, spleen and liver rather than in the bone marrow after transplantation of MISTRG mice with human CD34+ hematopoietic stem and progenitor cells. Due to their strategic location, human CD5+ ILCs could serve as blood-borne sentinels, ready to be recruited into the lung to respond to environmental challenges. This work emphasizes the uniqueness of human CD5+ ILCs in terms of their anatomical localization and developmental origin compared to well-studied CD5- ILCs.


Asunto(s)
Antígenos CD5/inmunología , Pulmón/inmunología , Linfocitos/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Movimiento Celular , Citocinas/inmunología , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunidad Innata , Masculino , Ratones , Persona de Mediana Edad , Bazo/inmunología
7.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836578

RESUMEN

Human adaptive-like "memory" CD56dimCD16+ natural killer (NK) cells in peripheral blood from cytomegalovirus-seropositive individuals have been extensively investigated in recent years and are currently explored as a treatment strategy for hematological cancers. However, treatment of solid tumors remains limited due to insufficient NK cell tumor infiltration, and it is unknown whether large expansions of adaptive-like NK cells that are equipped for tissue residency and tumor homing exist in peripheral tissues. Here, we show that human lung and blood contains adaptive-like CD56brightCD16- NK cells with hallmarks of tissue residency, including expression of CD49a. Expansions of adaptive-like lung tissue-resident NK (trNK) cells were found to be present independently of adaptive-like CD56dimCD16+ NK cells and to be hyperresponsive toward target cells. Together, our data demonstrate that phenotypically, functionally, and developmentally distinct subsets of adaptive-like NK cells exist in human lung and blood. Given their tissue-related character and hyperresponsiveness, human lung adaptive-like trNK cells might represent a suitable alternative for therapies targeting solid tumors.


Asunto(s)
Células Asesinas Naturales/inmunología , Pulmón/inmunología , Adaptación Fisiológica/inmunología , Citometría de Flujo , Humanos , Inmunofenotipificación , Integrina alfa1/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia
8.
Immunity ; 54(2): 259-275.e7, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33382972

RESUMEN

The study of human macrophages and their ontogeny is an important unresolved issue. Here, we use a humanized mouse model expressing human cytokines to dissect the development of lung macrophages from human hematopoiesis in vivo. Human CD34+ hematopoietic stem and progenitor cells (HSPCs) generated three macrophage populations, occupying separate anatomical niches in the lung. Intravascular cell labeling, cell transplantation, and fate-mapping studies established that classical CD14+ blood monocytes derived from HSPCs migrated into lung tissue and gave rise to human interstitial and alveolar macrophages. In contrast, non-classical CD16+ blood monocytes preferentially generated macrophages resident in the lung vasculature (pulmonary intravascular macrophages). Finally, single-cell RNA sequencing defined intermediate differentiation stages in human lung macrophage development from blood monocytes. This study identifies distinct developmental pathways from circulating monocytes to lung macrophages and reveals how cellular origin contributes to human macrophage identity, diversity, and localization in vivo.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Antígenos CD34/metabolismo , Biodiversidad , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Sangre Fetal/citología , Humanos , Receptores de Lipopolisacáridos/metabolismo , Pulmón/irrigación sanguínea , Receptores de IgG/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Nicho de Células Madre
9.
Sci Immunol ; 5(50)2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32826343

RESUMEN

Understanding innate immune responses in COVID-19 is important to decipher mechanisms of host responses and interpret disease pathogenesis. Natural killer (NK) cells are innate effector lymphocytes that respond to acute viral infections but might also contribute to immunopathology. Using 28-color flow cytometry, we here reveal strong NK cell activation across distinct subsets in peripheral blood of COVID-19 patients. This pattern was mirrored in scRNA-seq signatures of NK cells in bronchoalveolar lavage from COVID-19 patients. Unsupervised high-dimensional analysis of peripheral blood NK cells furthermore identified distinct NK cell immunotypes that were linked to disease severity. Hallmarks of these immunotypes were high expression of perforin, NKG2C, and Ksp37, reflecting increased presence of adaptive NK cells in circulation of patients with severe disease. Finally, arming of CD56bright NK cells was observed across COVID-19 disease states, driven by a defined protein-protein interaction network of inflammatory soluble factors. This study provides a detailed map of the NK cell activation landscape in COVID-19 disease.


Asunto(s)
Betacoronavirus/genética , Betacoronavirus/inmunología , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/inmunología , Células Asesinas Naturales/inmunología , Neumonía Viral/epidemiología , Neumonía Viral/inmunología , Índice de Severidad de la Enfermedad , Inmunidad Adaptativa , Antígeno CD56/metabolismo , COVID-19 , Infecciones por Coronavirus/sangre , Infecciones por Coronavirus/patología , Femenino , Citometría de Flujo/métodos , Humanos , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Pandemias , Fenotipo , Neumonía Viral/sangre , Neumonía Viral/patología , Reacción en Cadena de la Polimerasa , Estudios Prospectivos , Mapas de Interacción de Proteínas/inmunología , Receptores KIR/metabolismo , SARS-CoV-2 , Pruebas Serológicas , Suecia/epidemiología
10.
Nat Commun ; 10(1): 3841, 2019 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-31451696

RESUMEN

Human lung tissue-resident NK cells (trNK cells) are likely to play an important role in host responses towards viral infections, inflammatory conditions and cancer. However, detailed insights into these cells are still largely lacking. Here we show, using RNA sequencing and flow cytometry-based analyses, that subsets of human lung CD69+CD16- NK cells display hallmarks of tissue-residency, including high expression of CD49a, CD103, and ZNF683, and reduced expression of SELL, S1PR5, and KLF2/3. CD49a+CD16- NK cells are functionally competent, and produce IFN-γ, TNF, MIP-1ß, and GM-CSF. After stimulation with IL-15, they upregulate perforin, granzyme B, and Ki67 to a similar degree as CD49a-CD16- NK cells. Comparing datasets from trNK cells in human lung and bone marrow with tissue-resident memory CD8+ T cells identifies core genes co-regulated either by tissue-residency, cell-type or location. Together, our data indicate that human lung trNK cells have distinct features, likely regulating their function in barrier immunity.


Asunto(s)
Inmunidad Mucosa , Células Asesinas Naturales/metabolismo , Enfermedades Pulmonares/inmunología , Pulmón/citología , Transcriptoma/inmunología , Anciano , Anciano de 80 o más Años , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Conjuntos de Datos como Asunto , Femenino , Humanos , Células Asesinas Naturales/inmunología , Pulmón/inmunología , Pulmón/cirugía , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/cirugía , Masculino , Persona de Mediana Edad , Neumonectomía , RNA-Seq , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo
11.
Nat Commun ; 10(1): 3897, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467285

RESUMEN

Despite animal models showing that natural killer (NK) cells are important players in the early defense against many viral infections, the NK cell response is poorly understood in humans. Here we analyze the phenotype, temporal dynamics, regulation and trafficking of NK cells in a patient cohort with acute dengue virus infection. NK cells are robustly activated and proliferate during the first week after symptom debut. Increased IL-18 levels in plasma and in induced skin blisters of DENV-infected patients, as well as concomitant signaling downstream of the IL-18R, suggests an IL-18-dependent mechanism in driving the proliferative NK cell response. Responding NK cells have a less mature phenotype and a distinct chemokine-receptor imprint indicative of skin-homing. A corresponding NK cell subset can be localized to skin early during acute infection. These data provide evidence of an IL-18-driven NK cell proliferation and priming for skin-homing during an acute viral infection in humans.


Asunto(s)
Dengue/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Piel/inmunología , Animales , Antígenos CD , Antígenos de Diferenciación de Linfocitos T , Antígeno CD56/genética , Proliferación Celular , Virus del Dengue , Humanos , Interleucina-18/metabolismo , Lectinas Tipo C , Ratones , Fenotipo , Receptores CCR5 , Receptores CXCR3 , Receptores de Interleucina-18/metabolismo , Transducción de Señal
12.
Front Immunol ; 10: 1116, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31156653

RESUMEN

NK cells in the human lung respond to influenza A virus- (IAV-) infected target cells. However, the detailed functional capacity of human lung and peripheral blood NK cells remains to be determined in IAV and other respiratory viral infections. Here, we investigated the effects of IAV infection on human lung and peripheral blood NK cells in vitro and ex vivo following clinical infection. IAV infection of lung- and peripheral blood-derived mononuclear cells in vitro induced NK cell hyperresponsiveness to K562 target cells, including increased degranulation and cytokine production particularly in the CD56brightCD16- subset of NK cells. Furthermore, lung CD16- NK cells showed increased IAV-mediated but target cell-independent activation compared to CD16+ lung NK cells or total NK cells in peripheral blood. IAV infection rendered peripheral blood NK cells responsive toward the normally NK cell-resistant lung epithelial cell line A549, indicating that NK cell activation during IAV infection could contribute to killing of surrounding non-infected epithelial cells. In vivo, peripheral blood CD56dimCD16+ and CD56brightCD16- NK cells were primed during acute IAV infection, and a small subset of CD16-CD49a+CXCR3+ NK cells could be identified, with CD49a and CXCR3 potentially promoting homing to and tissue-retention in the lung during acute infection. Together, we show that IAV respiratory viral infections prime otherwise hyporesponsive lung NK cells, indicating that both CD16+ and CD16- NK cells including CD16-CD49a+ tissue-resident NK cells could contribute to host immunity but possibly also tissue damage in clinical IAV infection.


Asunto(s)
Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Pulmón/fisiología , Presentación de Antígeno , Circulación Sanguínea , Hiperreactividad Bronquial/metabolismo , Citotoxicidad Inmunológica , Humanos , Células K562 , Células Asesinas Naturales/inmunología , Activación de Linfocitos
13.
J Hepatol ; 71(2): 301-312, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31100314

RESUMEN

BACKGROUND & AIMS: Hepatitis delta virus (HDV) infection is the most severe form of viral hepatitis. Although HDV-associated liver disease is considered immune-mediated, adaptive immune responses against HDV are weak. Thus, the role of several other cell-mediated mechanisms such as those driven by mucosa-associated invariant T (MAIT) cells, a group of innate-like T cells highly enriched in the human liver, has not been extensively studied in clinical HDV infection. METHODS: MAIT cells from a sizeable cohort of patients with chronic HDV were analyzed ex vivo and in vitro after stimulation. Results were compared with MAIT cells from hepatitis B virus (HBV) monoinfected patients and healthy controls. RESULTS: Circulating MAIT cells were dramatically decreased in the peripheral blood of HDV-infected patients. Signs of decline were also observed in the liver. In contrast, only a modest decrease of circulating MAIT cells was noted in HBV monoinfection. Unsupervised high-dimensional analysis of residual circulating MAIT cells in chronic HDV infection revealed the appearance of a compound phenotype of CD38hiPD-1hiCD28loCD127loPLZFloEomesloHelioslo cells indicative of activation. Corroborating these results, MAIT cells exhibited a functionally impaired responsiveness. In parallel to MAIT cell loss, HDV-infected patients exhibited signs of monocyte activation and increased levels of proinflammatory cytokines IL-12 and IL-18. In vitro, IL-12 and IL-18 induced an activated MAIT cell phenotype similar to the one observed ex vivo in HDV-infected patients. These cytokines also promoted MAIT cell death, suggesting that they may contribute to MAIT cell activation and subsequent loss during HDV infection. CONCLUSIONS: These results suggest that chronic HDV infection engages the MAIT cell compartment causing activation, functional impairment, and subsequent progressive loss of MAIT cells as the HDV-associated liver disease progresses. LAY SUMMARY: Hepatitis delta virus (HDV) infection is the most severe form of viral hepatitis. We found that in patients with HDV, a subset of innate-like T cells called mucosa-associated invariant T cells (or MAIT cells), which are normally abundant in peripheral blood and the liver, are activated, functionally impaired and severely depleted.


Asunto(s)
Hepatitis D Crónica/inmunología , Virus de la Hepatitis Delta/fisiología , Activación de Linfocitos/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Adulto , Anciano , Linfocitos T CD8-positivos/inmunología , Estudios de Cohortes , Femenino , Células Hep G2 , Hepatitis D Crónica/virología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Interleucina-12/sangre , Interleucina-18/sangre , Hígado/patología , Masculino , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor/metabolismo , Fenotipo , Receptores de Antígenos de Linfocitos T/metabolismo , Adulto Joven
14.
PLoS One ; 13(9): e0203793, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30199551

RESUMEN

In the European Union (EU) millions of laboratory mice are used and killed for experimental and other scientific purposes each year. Although controversially discussed, the use of carbon dioxide (CO2) is still permitted for killing rodents according to the Directive 2010/63/EU. Within the scope of refinement, our aim was to investigate if isoflurane and sevoflurane are an appropriate alternative killing method to CO2 in mice. Different concentrations of CO2 (filling rates of 20%, 60%, 100%; CO2 20, 60, 100), isoflurane (Iso 2%, 5%) and sevoflurane (Sevo 4.8%, 8%) were compared in two mouse strains (NMRI, C57Bl/6J) using a broad spectrum of behavioral parameters, including the approach-avoidance test, and analyzing blood for stress parameters (glucose, adrenaline, noradrenaline). We focused in our study on the period from the beginning of the gas inlet to loss of consciousness, as during this period animals are able to perceive pain and distress. Our results show that only higher concentrations of CO2 (CO2 60, 100) and isoflurane (5%) induced surgical tolerance within 300 s in both strains, with CO2 100 being the fastest acting inhalant anesthetic. The potency of halogenated ethers depended on the mouse strain, with C57Bl/6J being more susceptible than NMRI mice. Behavioral analysis revealed no specific signs of distress, e. g. stress-induced grooming, and pain, i. e. audible vocalizations, for all inhalant gases. However, adrenaline and noradrenaline plasma concentrations were increased, especially in NMRI mice exposed to CO2 in high concentrations, whereas we did not observe such increase in animals exposed to isoflurane or sevoflurane. Escape latencies in the approach-avoidance test using C57Bl/6J mice did not differ between the three inhalant gases, however, some animals became recumbent during isoflurane and sevoflurane but not during CO2 exposure. The rise in catecholamine concentrations suggests that CO2 exposure might be linked to a higher stress response compared to isoflurane and sevoflurane exposure, although we did not observe a behavioral correlate for that. Follow-up studies investigating other fast-acting stress hormones and central anxiety circuits are needed to confirm our findings.


Asunto(s)
Anestésicos por Inhalación , Dióxido de Carbono , Eutanasia Animal/métodos , Isoflurano , Sevoflurano , Anestésicos por Inhalación/administración & dosificación , Anestésicos por Inhalación/efectos adversos , Animales , Animales de Laboratorio , Conducta Animal/efectos de los fármacos , Glucemia/metabolismo , Dióxido de Carbono/administración & dosificación , Dióxido de Carbono/efectos adversos , Epinefrina/sangre , Femenino , Isoflurano/administración & dosificación , Isoflurano/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Norepinefrina/sangre , Sevoflurano/administración & dosificación , Sevoflurano/efectos adversos , Especificidad de la Especie , Estrés Fisiológico/efectos de los fármacos
15.
J Allergy Clin Immunol ; 140(1): 318, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28528925
16.
Immunity ; 46(2): 287-300, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28214226

RESUMEN

Tissue-resident memory T (Trm) cells form a heterogeneous population that provides localized protection against pathogens. Here, we identify CD49a as a marker that differentiates CD8+ Trm cells on a compartmental and functional basis. In human skin epithelia, CD8+CD49a+ Trm cells produced interferon-γ, whereas CD8+CD49a- Trm cells produced interleukin-17 (IL-17). In addition, CD8+CD49a+ Trm cells from healthy skin rapidly induced the expression of the effector molecules perforin and granzyme B when stimulated with IL-15, thereby promoting a strong cytotoxic response. In skin from patients with vitiligo, where melanocytes are eradicated locally, CD8+CD49a+ Trm cells that constitutively expressed perforin and granzyme B accumulated both in the epidermis and dermis. Conversely, CD8+CD49a- Trm cells from psoriasis lesions predominantly generated IL-17 responses that promote local inflammation in this skin disease. Overall, CD49a expression delineates CD8+ Trm cell specialization in human epithelial barriers and correlates with the effector cell balance found in distinct inflammatory skin diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica/inmunología , Integrina alfa1/inmunología , Piel/inmunología , Subgrupos de Linfocitos T/inmunología , Separación Celular , Citometría de Flujo , Humanos , Memoria Inmunológica/inmunología , Integrina alfa1/biosíntesis , Activación de Linfocitos/inmunología , Microscopía Confocal , Psoriasis/inmunología , Vitíligo/inmunología
17.
J Allergy Clin Immunol ; 139(4): 1321-1330.e4, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27670241

RESUMEN

BACKGROUND: In contrast to the extensive knowledge about human natural killer (NK) cells in peripheral blood, relatively little is known about NK cells in the human lung. Knowledge about the composition, differentiation, and function of human lung NK cells is critical to better understand their role in diseases affecting the lung, including asthma, chronic obstructive pulmonary disease, infections, and cancer. OBJECTIVE: We sought to analyze and compare the phenotypic and functional characteristics of NK cells in the human lung and peripheral blood at the single-cell level. METHODS: NK cells in human lung tissue and matched peripheral blood from 132 subjects were analyzed by using 16-color flow cytometry and confocal microscopy. RESULTS: CD56dimCD16+ NK cells made up the vast majority of NK cells in human lungs, had a more differentiated phenotype, and more frequently expressed educating killer cell immunoglobulin-like receptors compared with NK cells in peripheral blood. Despite this, human lung NK cells were hyporesponsive toward target cell stimulation, even after priming with IFN-α. Furthermore, we detected a small subset of NK cells expressing CD69, a marker of tissue residency. These CD69+ NK cells in the lung consisted predominantly of immature CD56brightCD16- NK cells and less differentiated CD56dimCD16+ NK cells. CONCLUSION: Here, we characterize the major NK cell populations in the human lung. Our data suggest a model in which the majority of NK cells in the human lung dynamically move between blood and the lung rather than residing in the lung as bona fide tissue-resident CD69+ NK cells.


Asunto(s)
Células Asesinas Naturales/citología , Pulmón/citología , Pulmón/inmunología , Subgrupos Linfocitarios/citología , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígeno CD56/inmunología , Diferenciación Celular/inmunología , Citometría de Flujo , Humanos , Células Asesinas Naturales/inmunología , Lectinas Tipo C/inmunología , Subgrupos Linfocitarios/inmunología , Microscopía Confocal
18.
J Immunol ; 197(8): 3069-3075, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27591320

RESUMEN

Amniotic fluid (AF) surrounds the growing fetus, and cells derived from AF are commonly used for diagnosis of genetic diseases. Intra-amniotic infections are strongly linked to preterm birth, which is the leading cause of perinatal mortality worldwide. Surprisingly little is known, however, about mature hematopoietic cells in AF, which could potentially be involved in immune responses during pregnancy. In this study, we show that the dominating population of viable CD45+ cells in AF is represented by a subset of fetal CD103+ group 3 innate lymphoid cells (ILCs) producing high levels of IL-17 and TNF. Fetal CD103+ ILC3s could also be detected at high frequency in second-trimester mucosal tissues (e.g., the intestine and lung). Taken together, our data indicate that CD103+ ILC3s accumulate with gestation in the fetal intestine and subsequently egress to the AF. The dominance of ILC3s producing IL-17 and TNF in AF suggests that they could be involved in controlling intra-amniotic infections and inflammation and as such could be important players in regulating subsequent premature birth.


Asunto(s)
Líquido Amniótico/inmunología , Mucosa Intestinal/inmunología , Subgrupos Linfocitarios/inmunología , Linfocitos/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Mucosa Respiratoria/inmunología , Antígenos CD/metabolismo , Células Cultivadas , Femenino , Feto , Humanos , Inmunidad Innata , Recién Nacido , Cadenas alfa de Integrinas/metabolismo , Interleucina-17/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Embarazo , Segundo Trimestre del Embarazo , Factor de Necrosis Tumoral alfa/metabolismo
19.
Animals (Basel) ; 6(9)2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-27563926

RESUMEN

Millions of laboratory animals are killed each year worldwide. There is an ethical, and in many countries also a legal, imperative to ensure those deaths cause minimal suffering. However, there is a lack of consensus regarding what methods of killing are humane for many species and stages of development. In 2013, an international group of researchers and stakeholders met at Newcastle University, United Kingdom to discuss the latest research and which methods could currently be considered most humane for the most commonly used laboratory species (mice, rats and zebrafish). They also discussed factors to consider when making decisions about appropriate techniques for particular species and projects, and priorities for further research. This report summarises the research findings and discussions, with recommendations to help inform good practice for humane killing.

20.
J Immunol ; 195(7): 3262-72, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26283480

RESUMEN

NK cells play an important role in the defense against viral infections. However, little is known about the regulation of NK cell responses during the first days of acute viral infections in humans. In this study, we used the live attenuated yellow fever virus (YFV) vaccine 17D as a human in vivo model to study the temporal dynamics and regulation of NK cell responses in an acute viral infection. YFV induced a robust NK cell response in vivo, with an early activation and peak in NK cell function at day 6, followed by a delayed peak in Ki67 expression, which was indicative of proliferation, at day 10. The in vivo NK cell response correlated positively with plasma type I/III IFN levels at day 6, as well as with the viral load. YFV induced an increased functional responsiveness to IL-12 and IL-18, as well as to K562 cells, indicating that the NK cells were primed in vivo. The NK cell responses were associated primarily with the stage of differentiation, because the magnitude of induced Ki67 and CD69 expression was distinctly higher in CD57(-) NK cells. In contrast, NK cells expressing self- and nonself-HLA class I-binding inhibitory killer cell Ig-like receptors contributed, to a similar degree, to the response. Taken together, our results indicate that NK cells are primed by type I/III IFN in vivo early after YFV infection and that their response is governed primarily by the differentiation stage, independently of killer cell Ig-like receptor/HLA class I-mediated inhibition or education.


Asunto(s)
Interferón Tipo I/inmunología , Células Asesinas Naturales/inmunología , Receptores KIR/inmunología , Vacuna contra la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/inmunología , Adulto , Anticuerpos Neutralizantes/inmunología , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Linfocitos B/inmunología , Antígenos CD57/metabolismo , Diferenciación Celular/inmunología , Proliferación Celular , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interferón Tipo I/sangre , Subunidad p35 de la Interleucina-12/inmunología , Interleucina-18/inmunología , Células K562 , Antígeno Ki-67/biosíntesis , Células Asesinas Naturales/citología , Lectinas Tipo C/biosíntesis , Activación de Linfocitos/inmunología , Persona de Mediana Edad , Linfocitos T/inmunología , Vacunas Atenuadas/inmunología , Carga Viral/inmunología , Vacunas Virales/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA