Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Nat Immunol ; 19(9): 954-962, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30127438

RESUMEN

Controlling the balance between immunity and immunopathology is crucial for host resistance to pathogens. After infection, activation of the hypothalamic-pituitary-adrenal (HPA) axis leads to the production of glucocorticoids. However, the pleiotropic effects of these steroid hormones make it difficult to delineate their precise role(s) in vivo. Here we found that the regulation of natural killer (NK) cell function by the glucocorticoid receptor (GR) was required for host survival after infection with mouse cytomegalovirus (MCMV). Mechanistically, endogenous glucocorticoids produced shortly after infection induced selective and tissue-specific expression of the checkpoint receptor PD-1 on NK cells. This glucocorticoid-PD-1 pathway limited production of the cytokine IFN-γ by spleen NK cells, which prevented immunopathology. Notably, this regulation did not compromise viral clearance. Thus, the fine tuning of NK cell functions by the HPA axis preserved tissue integrity without impairing pathogen elimination, which reveals a novel aspect of neuroimmune regulation.


Asunto(s)
Glucocorticoides/metabolismo , Infecciones por Herpesviridae/inmunología , Células Asesinas Naturales/fisiología , Muromegalovirus/fisiología , Receptor de Muerte Celular Programada 1/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Células Cultivadas , Femenino , Sistema Hipotálamo-Hipofisario , Inmunidad Innata , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación , Especificidad de Órganos , Sistema Hipófiso-Suprarrenal , Receptores de Glucocorticoides/genética , Transducción de Señal , Carga Viral
2.
Nature ; 594(7861): 94-99, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34012116

RESUMEN

Inflammation is a defence response to tissue damage that requires tight regulation in order to prevent impaired healing. Tissue-resident macrophages have a key role in tissue repair1, but the precise molecular mechanisms that regulate the balance between inflammatory and pro-repair macrophage responses during healing remain poorly understood. Here we demonstrate a major role for sensory neurons in promoting the tissue-repair function of macrophages. In a sunburn-like model of skin damage in mice, the conditional ablation of sensory neurons expressing the Gαi-interacting protein (GINIP) results in defective tissue regeneration and in dermal fibrosis. Elucidation of the underlying molecular mechanisms revealed a crucial role for the neuropeptide TAFA4, which is produced in the skin by C-low threshold mechanoreceptors-a subset of GINIP+ neurons. TAFA4 modulates the inflammatory profile of macrophages directly in vitro. In vivo studies in Tafa4-deficient mice revealed that TAFA4 promotes the production of IL-10 by dermal macrophages after UV-induced skin damage. This TAFA4-IL-10 axis also ensures the survival and maintenance of IL-10+TIM4+ dermal macrophages, reducing skin inflammation and promoting tissue regeneration. These results reveal a neuroimmune regulatory pathway driven by the neuropeptide TAFA4 that promotes the anti-inflammatory functions of macrophages and prevents fibrosis after tissue damage, and could lead to new therapeutic perspectives for inflammatory diseases.


Asunto(s)
Citocinas/metabolismo , Macrófagos/metabolismo , Regeneración , Células Receptoras Sensoriales/metabolismo , Cicatrización de Heridas , Animales , Supervivencia Celular , Citocinas/deficiencia , Modelos Animales de Enfermedad , Femenino , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Fibrosis/prevención & control , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Interleucina-10/biosíntesis , Interleucina-10/metabolismo , Macrófagos/efectos de la radiación , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Receptoras Sensoriales/efectos de la radiación , Piel/patología , Piel/efectos de la radiación , Quemadura Solar/complicaciones , Quemadura Solar/etiología , Quemadura Solar/metabolismo , Quemadura Solar/patología , Rayos Ultravioleta/efectos adversos
3.
Gut ; 72(6): 1115-1128, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36175116

RESUMEN

OBJECTIVE: In the management of patients with IBD, there is a need to identify prognostic markers and druggable biological pathways to improve mucosal repair and probe the efficacy of tumour necrosis factor alpha biologics. Vnn1 is a pantetheinase that degrades pantetheine to pantothenate (vitamin B5, a precursor of coenzyme A (CoA) biosynthesis) and cysteamine. Vnn1 is overexpressed by inflamed colonocytes. We investigated its contribution to the tolerance of the intestinal mucosa to colitis-induced injury. DESIGN: We performed an RNA sequencing study on colon biopsy samples from patients with IBD stratified according to clinical severity and modalities of treatment. We generated the VIVA mouse transgenic model, which specifically overexpresses Vnn1 on intestinal epithelial cells and explored its susceptibility to colitis. We developed a pharmacological mimicry of Vnn1 overexpression by administration of Vnn1 derivatives. RESULTS: VNN1 overexpression on colonocytes correlates with IBD severity. VIVA mice are resistant to experimentally induced colitis. The pantetheinase activity of Vnn1 is cytoprotective in colon: it enhances CoA regeneration and metabolic adaptation of colonocytes; it favours microbiota-dependent production of short chain fatty acids and mostly butyrate, shown to regulate mucosal energetics and to be reduced in patients with IBD. This prohealing phenotype is recapitulated by treating control mice with the substrate (pantethine) or the products of pantetheinase activity prior to induction of colitis. In severe IBD, the protection conferred by the high induction of VNN1 might be compromised because its enzymatic activity may be limited by lack of available substrates. In addition, we identify the elevation of indoxyl sulfate in urine as a biomarker of Vnn1 overexpression, also detected in patients with IBD. CONCLUSION: The induction of Vnn1/VNN1 during colitis in mouse and human is a compensatory mechanism to reinforce the mucosal barrier. Therefore, enhancement of vitamin B5-driven metabolism should improve mucosal healing and might increase the efficacy of anti-inflammatory therapy.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Humanos , Ratones , Animales , Colitis/metabolismo , Colon/patología , Mucosa Intestinal/metabolismo , Enfermedades Inflamatorias del Intestino/genética , Ácidos Grasos Volátiles/metabolismo , Vitaminas , Sulfato de Dextran , Modelos Animales de Enfermedad
4.
Cell Mol Life Sci ; 79(7): 355, 2022 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-35678896

RESUMEN

Thymically-derived Foxp3+ regulatory T cells (Treg) critically control immunological tolerance. These cells are generated in the medulla through high affinity interactions with medullary thymic epithelial cells (mTEC) expressing the Autoimmune regulator (Aire). Recent advances have revealed that thymic Treg contain not only developing but also recirculating cells from the periphery. Although Aire is implicated in the generation of Foxp3+ Treg, its role in the biology of recirculating Treg remains elusive. Here, we show that Aire regulates the suppressive signature of recirculating Treg independently of the remodeling of the medullary 3D organization throughout life where Treg reside. Accordingly, the adoptive transfer of peripheral Foxp3+ Treg in AireKO recipients led to an impaired suppressive signature upon their entry into the thymus. Furthermore, recirculating Treg from AireKO mice failed to attenuate the severity of multiorgan autoimmunity, demonstrating that their suppressive function is altered. Using bone marrow chimeras, we reveal that mTEC-specific expression of Aire controls the suppressive signature of recirculating Treg. Finally, mature mTEC lacking Aire were inefficient in stimulating peripheral Treg both in polyclonal and antigen-specific co-culture assays. Overall, this study demonstrates that Aire confers to mTEC the ability to restimulate recirculating Treg, unravelling a novel function for this master regulator in Treg biology.


Asunto(s)
Tolerancia Inmunológica , Linfocitos T Reguladores , Animales , Autoinmunidad , Células Epiteliales/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Ratones , Timo
5.
Nat Immunol ; 10(1): 75-82, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19029904

RESUMEN

NKp46+CD3- natural killer lymphocytes isolated from blood, lymphoid organs, lung, liver and uterus can produce granule-dependent cytotoxicity and interferon-gamma. Here we identify in dermis, gut lamina propria and cryptopatches distinct populations of NKp46+CD3- cells with a diminished capacity to degranulate and produce interferon-gamma. In the gut, expression of the transcription factor RORgammat, which is involved in the development of lymphoid tissue-inducer cells, defined a previously unknown subset of NKp46+CD3- lymphocytes. Unlike RORgammat- lamina propria and dermis natural killer cells, gut RORgammat+NKp46+ cells produced interleukin 22. Our data show that lymphoid tissue-inducer cells and natural killer cells shared unanticipated similarities and emphasize the heterogeneity of NKp46+CD3- cells in innate immunity, lymphoid organization and local tissue repair.


Asunto(s)
Dermis/inmunología , Mucosa Intestinal/inmunología , Células T Asesinas Naturales/inmunología , Receptores de Ácido Retinoico/fisiología , Receptores de Hormona Tiroidea/fisiología , Factores de Transcripción/fisiología , Animales , Complejo CD3/metabolismo , División Celular , Humanos , Interferón gamma/biosíntesis , Interleucinas/biosíntesis , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Receptor 1 Gatillante de la Citotoxidad Natural/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Ganglios Linfáticos Agregados/inmunología , Receptores de Ácido Retinoico/genética , Receptores de Hormona Tiroidea/genética , Factores de Transcripción/genética , Interleucina-22
6.
Clin Infect Dis ; 70(11): 2405-2412, 2020 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31309973

RESUMEN

BACKGROUND: Etiological diagnosis is a key to therapeutic adaptation and improved prognosis, particularly for infections such as endocarditis. In blood culture-negative endocarditis (BCNE), 22% of cases remain undiagnosed despite an updated comprehensive syndromic approach. This prompted us to develop a new diagnostic approach. METHODS: Eleven valves from 10 BCNE patients were analyzed using a method that combines human RNA bait-depletion with phi29 DNA polymerase-based multiple displacement amplification and shotgun DNA sequencing. An additional case in which a microbe was serendipitously visualized by immunofluorescence was analyzed using the same method, but after laser capture microdissection. RESULTS: Background DNA prevented any diagnosis in cases analyzed without microdissection because the majority of sequences were contaminants. Moraxella sequences were dramatically enriched in the stained microdissected region of the additional case. A consensus genome sequence of 2.4 Mbp covering more than 94% of the Moraxella osloensis KSH reference genome was reconstructed with 234X average coverage. Several antibiotic-resistance genes were observed. Etiological diagnosis was confirmed using Western blot and specific polymerase chain reaction with sequencing on a different valve sample. CONCLUSIONS: Microdissection could be a key to the metagenomic diagnosis of infectious diseases when a microbe is visualized but remains unidentified despite an updated optimal approach. Moraxella osloensis should be tested in blood culture-negative endocarditis.


Asunto(s)
Endocarditis Bacteriana , Endocarditis , Cultivo de Sangre , Endocarditis/diagnóstico , Endocarditis Bacteriana/diagnóstico , Humanos , Metagenómica , Moraxella
7.
J Neuroinflammation ; 16(1): 191, 2019 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-31660979

RESUMEN

BACKGROUND: Targeting angiogenesis has been and continues to be an attractive therapeutic modality in glioblastoma (GBM) patients. However, GBM rapidly becomes refractory to anti-VEGF therapies. Myeloid cell infiltration is an important determinant of tumor progression. Given that VEGF is a modulator of the innate immune response we sought to analyze the dynamics of this response in a mouse model of GBM undergoing anti-VEGF therapy. METHODS: We grafted GL261-DsRed cells in transgenic Thy1-CFP//LysM-EGFP//CD11c-EYFP reporter mice. We combined recurrent spectral two-photon imaging with multiparametric cytometry, immunostaining, and brain clearing to characterize at two critical stages of tumor development (day 21 and day 28 after tumor grafting) the nature and spatial distribution of the innate response in control and bevacizumab-treated mice. RESULTS: We report that at an early stage (21 day), VEGF blockade has a detectable effect on the number of microglial cells but only a mild effect on the number of infiltrating myeloid cells. At a later stage (day 28), the treatment resulted in a specific adjustment of dendritic cell subsets. In treated mice, the number of monocytes and their monocyte-derived dendritic cells (moDC) progeny was increased by approximately twofold compared to untreated mice. In agreement, by in vivo quantitative imaging, we observed that treatment increased the number of LysM-EGFP cells traveling in tumor blood vessels and doubled the densities of both infiltrated LysM-EGFP monocytes and double-labeled EGFP/EYFP moDC. The treatment also led to an increased density of conventional cDCs2 subset together with a decrease of cDCs1 subset, necessary for the development of anti-tumor immunity. Finally, we describe differential spatial cell distributions and two immune cell-traveling routes into the brain. LysM-EGFP cells distributed as a gradient from the meninges towards the tumor whereas CD11c-EYFP/MHCII+ cells were located in the basal area of the tumor. Brain clearing also revealed a flow of CD11c-EYFP cells following the corpus callosum. CONCLUSION: We uncovered new features in the dynamics of innate immune cells in GBM-bearing mice and deciphered precisely the key populations, i.e., DC subsets controlling immune responses, that are affected by VEGF blockade. Since despite differences, human pathogenesis presents similarities with our mouse model, the data provide new insights into the effect of bevacizumab at the cellular level.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/patología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Carga Tumoral/efectos de los fármacos , Carga Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Biochim Biophys Acta ; 1862(4): 662-669, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26850476

RESUMEN

BACKGROUND AND AIMS: Pretreatment with clofibrate, a peroxisome proliferator-activated receptor alpha (PPARa) agonist, protects mice from acetaminophen (APAP) injury. Protection is not due to alterations in APAP metabolism and is dependent on PPARa expression. Gene array analysis revealed that mice receiving clofibrate have enhanced hepatic Vanin-1 (Vnn1) gene expression, a response that is also PPARa dependent. METHODS: We examined the role of Vnn1 by comparing the responses of Vnn1 knockout and wild-type mice following APAP hepatotoxicity. APAP metabolism, hepatotoxicity, and compensatory hepatocyte proliferation and immune responses were assessed. RESULTS: Vnn1 knockout mice are more susceptible to APAP hepatotoxicity despite no differences in hepatic glutathione content, gene expression of APAP metabolizing enzymes, or hepatic capacity to bioactivate or detoxify APAP ex vivo. Together, these data strongly suggest that the susceptibility of Vnn1 knockout mice is not due to differences in APAP metabolism. Immunochemistry revealed a lack of proliferating cell nuclear antigen-positive hepatocytes and F4/80-positive macrophages in and around areas of centrilobular necrosis in APAP-treated Vnn1 knockouts. Hepatic gene induction of pro-inflammatory cytokines was either significantly reduced or completely blunted in these mice. This was correlated with a reduction in early recruitment of cells positive for granulocyte differentiation antigen 1 or integrin alpha M. Heightened toxicity was also observed in CCl4 and ConA hepatitis models in the absence of Vnn1. CONCLUSIONS: These results indicate that mice lacking Vnn1 have deficiencies in compensatory repair and immune responses following toxic APAP exposure and that these mechanisms may contribute to the enhanced hepatotoxicity seen.


Asunto(s)
Acetaminofén/efectos adversos , Amidohidrolasas/deficiencia , Proliferación Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Hígado/inmunología , Acetaminofén/farmacología , Amidohidrolasas/inmunología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Clofibrato/farmacología , Proteínas Ligadas a GPI/deficiencia , Proteínas Ligadas a GPI/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Hepatocitos/inmunología , Hepatocitos/patología , Hígado/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , PPAR alfa/genética , PPAR alfa/inmunología
9.
PLoS Pathog ; 11(5): e1004897, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25954804

RESUMEN

In mice, plasmacytoid dendritic cells (pDC) and natural killer (NK) cells both contribute to resistance to systemic infections with herpes viruses including mouse Cytomegalovirus (MCMV). pDCs are the major source of type I IFN (IFN-I) during MCMV infection. This response requires pDC-intrinsic MyD88-dependent signaling by Toll-Like Receptors 7 and 9. Provided that they express appropriate recognition receptors such as Ly49H, NK cells can directly sense and kill MCMV-infected cells. The loss of any one of these responses increases susceptibility to infection. However, the relative importance of these antiviral immune responses and how they are related remain unclear. In humans, while IFN-I responses are essential, MyD88 is dispensable for antiviral immunity. Hence, a higher redundancy has been proposed in the mechanisms promoting protective immune responses against systemic infections by herpes viruses during natural infections in humans. It has been assumed, but not proven, that mice fail to mount protective MyD88-independent IFN-I responses. In humans, the mechanism that compensates MyD88 deficiency has not been elucidated. To address these issues, we compared resistance to MCMV infection and immune responses between mouse strains deficient for MyD88, the IFN-I receptor and/or Ly49H. We show that selective depletion of pDC or genetic deficiencies for MyD88 or TLR9 drastically decreased production of IFN-I, but not the protective antiviral responses. Moreover, MyD88, but not IFN-I receptor, deficiency could largely be compensated by Ly49H-mediated antiviral NK cell responses. Thus, contrary to the current dogma but consistent with the situation in humans, we conclude that, in mice, in our experimental settings, MyD88 is redundant for IFN-I responses and overall defense against a systemic herpes virus infection. Moreover, we identified direct NK cell sensing of infected cells as one mechanism able to compensate for MyD88 deficiency in mice. Similar mechanisms likely contribute to protect MyD88- or IRAK4-deficient patients from viral infections.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Células Asesinas Naturales/inmunología , Muromegalovirus/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor de Interferón alfa y beta/agonistas , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Infecciones por Herpesviridae/sangre , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Inmunidad Innata , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/virología , Interferón Tipo I/sangre , Interleucina-12/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/virología , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Mutantes , Muromegalovirus/fisiología , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Subfamilia A de Receptores Similares a Lectina de Células NK/deficiencia , Subfamilia A de Receptores Similares a Lectina de Células NK/genética , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Enfermedades de Inmunodeficiencia Primaria , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , Organismos Libres de Patógenos Específicos , Bazo/inmunología , Bazo/metabolismo , Bazo/virología , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
10.
Proc Natl Acad Sci U S A ; 111(30): 11115-20, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25024217

RESUMEN

The transcription factor NF-κB is central to inflammatory signaling and activation of innate and adaptive immune responses. Activation of the NF-κB pathway is tightly controlled by several negative feedback mechanisms, including A20, an ubiquitin-modifying enzyme encoded by the tnfaip3 gene. Mice with selective deletion of A20 in myeloid, dendritic, or B cells recapitulate some human inflammatory pathology. As we observed high expression of A20 transcripts in dysfunctional CD8 T cells in an autochthonous melanoma, we analyzed the role of A20 in regulation of CD8 T-cell functions, using mice in which A20 was selectively deleted in mature conventional T cells. These mice developed lymphadenopathy and some organ infiltration by T cells but no splenomegaly and no detectable pathology. A20-deleted CD8 T cells had increased sensitivity to antigen stimulation with production of large amounts of IL-2 and IFNγ, correlated with sustained nuclear expression of NF-κB components reticuloendotheliosis oncogene c-Rel and p65. Overexpression of A20 by retroviral transduction of CD8 T cells dampened their intratumor accumulation and antitumor activity. In contrast, relief from the A20 brake in NF-κB activation in adoptively transferred antitumor CD8 T cells led to improved control of melanoma growth. Tumor-infiltrating A20-deleted CD8 T cells had enhanced production of IFNγ and TNFα and reduced expression of the inhibitory receptor programmed cell death 1. As manipulation of A20 expression in CD8 T cells did not result in pathologic manifestations in the mice, we propose it as a candidate to be targeted to increase antitumor efficiency of adoptive T-cell immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Cisteína Endopeptidasas/inmunología , Inmunidad Celular , Péptidos y Proteínas de Señalización Intracelular/inmunología , Melanoma/inmunología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/patología , Cisteína Endopeptidasas/genética , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Melanoma/genética , Melanoma/patología , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
11.
Am J Pathol ; 185(11): 3039-52, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26343328

RESUMEN

Tissue pantetheinase, encoded by the VNN1 gene, regulates response to stress, and previous studies have shown that VNN genes contribute to the susceptibility to malaria. Herein, we evaluated the role of pantetheinase on erythrocyte homeostasis and on the development of malaria in patients and in a new mouse model of pantetheinase insufficiency. Patients with cerebral malaria have significantly reduced levels of serum pantetheinase activity (PA). In mouse, we show that a reduction in serum PA predisposes to severe malaria, including cerebral malaria and severe anemia. Therefore, scoring pantetheinase in serum may serve as a severity marker in malaria infection. This disease triggers an acute stress in erythrocytes, which enhances cytoadherence and hemolysis. We speculated that serum pantetheinase might contribute to erythrocyte resistance to stress under homeostatic conditions. We show that mutant mice with a reduced serum PA are anemic and prone to phenylhydrazine-induced anemia. A cytofluorometric and spectroscopic analysis documented an increased frequency of erythrocytes with an autofluorescent aging phenotype. This is associated with an enhanced oxidative stress and shear stress-induced hemolysis. Red blood cell transfer and bone marrow chimera experiments show that the aging phenotype is not cell intrinsic but conferred by the environment, leading to a shortening of red blood cell half-life. Therefore, serum pantetheinase level regulates erythrocyte life span and modulates the risk of developing complicated malaria.


Asunto(s)
Amidohidrolasas/sangre , Eritrocitos/fisiología , Malaria/fisiopatología , Adolescente , Adulto , Amidohidrolasas/metabolismo , Anemia , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Proteínas Ligadas a GPI/sangre , Proteínas Ligadas a GPI/metabolismo , Homeostasis , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Adulto Joven
12.
Blood ; 123(22): 3462-5, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24677543

RESUMEN

Follicular lymphoma (FL) is a B-cell neoplasm resulting from the transformation of germinal center (GC) B cells. Although t(14;18) and ectopic B-cell lymphoma 2 (BCL2) expression constitute the genetic hallmark of FL, t(14;18)(pos) B cells bearing genotypic and phenotypic features of FL cells can be found in the blood of most healthy individuals. Nevertheless, the localization of these FL-like cells (FLLCs) in nonmalignant GC-rich tissues and the functional consequences of BCL2 overexpression have not been evaluated thus far. Among 85 reactive lymph node (RLN) samples, 14% were found to contain high levels of t(14;18) by quantitative polymerase chain reaction. In t(14;18)(hi) RLNs, CD20(pos)BCL2(pos)CD10(pos) FLLCs consistently accumulated within the GC, essentially as nonproliferative CXCR4(neg) centrocytes. Moreover, they displayed a reduced response to proliferative stimuli in vitro. Altogether, our findings provide new insights into in situ FLLC functional properties and suggest that these cells have not acquired the ultimate genetic events leading to FL transformation.


Asunto(s)
Cromosomas Humanos Par 14 , Cromosomas Humanos Par 18 , Linfoma de Células B/genética , Linfoma Folicular/genética , Translocación Genética , Adulto , Anciano , Antígenos CD20/genética , Antígenos CD20/metabolismo , Transformación Celular Neoplásica/genética , Femenino , Centro Germinal/metabolismo , Centro Germinal/patología , Humanos , Masculino , Persona de Mediana Edad , Neprilisina/genética , Neprilisina/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
13.
Immunology ; 145(1): 24-33, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25367158

RESUMEN

CD8 T cells contribute to long-term protection against Listeria monocytogenes infection by differentiating into memory T cells. These rapidly respond to antigen or inflammation upon secondary infection. In this study we used CD8 T cells from OT1 mice and CD4 T cells from OT2 mice expressing a fluorescent chimeric granzyme (GZMB-Tom) protein to monitor the primary response to infection with ovalbumin-expressing L. monocytogenes (Lm-OVA). We show that, unlike poorly responding CD4 T cells, CD8 T cells readily proliferated and expressed high levels of GZMB-Tom as early as 2 days after infection. FACS analysis showed GZMB-Tom expression in undivided CD8 T cells, with its level increasing over one to four divisions. OT1 T cells were visualized in the T-cell zone by confocal microscopy. This showed GZMB-Tom-containing granules oriented towards MHCII-positive cells. Twenty hours later, most OT1 T cells had divided but their level of GZMB-Tom expression was reduced. Recently divided OT1 cells failed to express GZMB-Tom. Fourteen hours after secondary infection, GZMB-Tom was re-expressed in memory OT1 T cells responding either to Lm-OVA or L. monocytogenes. Differences in the activation phenotype and in the splenic distribution of OT1 T cells were observed, depending on the challenge. Notably, OTI T cells with polarized granules were only observed after challenge with cognate antigen. This work showed that the GZMB-Tom knock-in mice in which GZMB-Tom faithfully reproduced GZMB expression, provide useful tools to dissect mechanisms leading to the development of anti-bacterial effector and memory CD8 T cells and reactivation of the memory response to cognate antigen or inflammatory signals.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Regulación Enzimológica de la Expresión Génica/inmunología , Granzimas/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Regulación Enzimológica de la Expresión Génica/genética , Granzimas/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Listeriosis/genética , Listeriosis/patología , Ratones , Ratones Transgénicos , Transducción de Señal/genética , Transducción de Señal/inmunología , Factores de Tiempo
14.
J Immunol ; 190(11): 5526-33, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23616571

RESUMEN

Galectin-1 (GAL1) is an S-type lectin with multiple functions, including the control of B cell homeostasis. GAL1 expression was reported to be under the control of the plasma cell master regulator BLIMP-1. GAL1 was detected at the protein level in LPS-stimulated B cells and was shown to promote Ig secretion in vitro. However, the pattern of GAL1 expression and function of GAL1 in B cells in vivo are still unclear. In this study, we show that, among B cells, GAL1 is only expressed by differentiating plasma cells following T-dependent or T-independent immunization. Using GAL1-deficient mice we demonstrate that GAL1 expression is required for the maintenance of Ag-specific Ig titers and Ab-secreting cell numbers. Using an in vitro differentiation assay we find that GAL1-deficient plasmablasts can develop normally but die rapidly, through caspase 8 activation, under serum starvation-induced death conditions. TUNEL assays show that in vivo-generated GAL1-deficient plasma cells exhibit an increased sensitivity to apoptosis. Taken together, our data indicate that endogenous GAL1 supports plasma cell survival and participates in the regulation of the humoral immune response.


Asunto(s)
Galectina 1/metabolismo , Homeostasis/inmunología , Inmunidad Humoral , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Animales , Antígenos/inmunología , Apoptosis/genética , Apoptosis/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Galectina 1/genética , Homeostasis/genética , Inmunidad Humoral/genética , Inmunoglobulina G/sangre , Inmunofenotipificación , Recuento de Linfocitos , Ratones , Ratones Noqueados , Células Plasmáticas/citología , Bazo/inmunología , Bazo/metabolismo
15.
Proc Natl Acad Sci U S A ; 109(8): 3006-11, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22315398

RESUMEN

In response to inflammatory stimulation, dendritic cells (DCs) have a remarkable pattern of differentiation that exhibits specific mechanisms to control the immune response. Here we show that in response to polyriboinosinic:polyribocytidylic acid (pI:C), DCs mount a specific integrated stress response during which the transcription factor ATF4 and the growth arrest and DNA damage-inducible protein 34 (GADD34/Ppp1r15a), a phosphatase 1 (PP1) cofactor, are expressed. In agreement with increased GADD34 levels, an extensive dephosphorylation of the translation initiation factor eIF2α was observed during DC activation. Unexpectedly, although DCs display an unusual resistance to protein synthesis inhibition induced in response to cytosolic dsRNA, GADD34 expression did not have a major impact on protein synthesis. GADD34, however, was shown to be required for normal cytokine production both in vitro and in vivo. These observations have important implications in linking further pathogen detection with the integrated stress response pathways.


Asunto(s)
Citocinas/biosíntesis , Células Dendríticas/efectos de los fármacos , Células Dendríticas/enzimología , Poli I-C/farmacología , Proteína Fosfatasa 1/metabolismo , Subunidades de Proteína/metabolismo , Factor de Transcripción Activador 4/metabolismo , Animales , Citosol/efectos de los fármacos , Citosol/metabolismo , Activación Enzimática/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Interferón beta/metabolismo , Ratones , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , eIF-2 Quinasa/metabolismo
16.
Genes Chromosomes Cancer ; 53(1): 52-66, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24249258

RESUMEN

MYC is a potent oncogene involved in ∼70% of human cancers, inducing tumorigenesis with high penetrance and short latency in experimental transgenic models. Accordingly, MYC is recognized as a major driver of T-cell acute lymphoblastic leukemia (T-ALL) in human and zebrafish/mouse models, and uncovering the context by which MYC-mediated malignant transformation initiates and develops remains a considerable challenge. Because MYC is a very complex oncogene, highly dependent on the microenvironment and cell-intrinsic context, we generated transgenic mice (tgMyc(spo)) in which ectopic Myc activation occurs sporadically (<10(-6) thymocytes) within otherwise normal thymic environment, thereby mimicking the unicellular context in which oncogenic alterations initiate human tumors. We show that while Myc(+) clones in tgMyc(spo) mice develop and initially proliferate in thymus and the periphery, no tumor or clonal expansion progress in aging mice (n = 130), suggesting an unexpectedly low ability of Myc to initiate efficient tumorigenesis. Furthermore, to determine the relevance of this observation in human pathogenesis we analyzed a human T-ALL case at diagnosis and relapse using the molecular stigmata of V(D)J recombination as markers of malignant progression; we similarly demonstrate that despite the occurrence of TAL1 and MYC translocations in early thymocyte ontogeny, subsequent oncogenic alterations were required to drive oncogenesis. Altogether, our data suggest that although central to T-ALL, MYC overexpression per se is inefficient in triggering the cascade of events leading to malignant transformation.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Genes myc/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Animales , Crisis Blástica/genética , Crisis Blástica/patología , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Transgénicos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Recurrencia , Translocación Genética , Recombinación V(D)J
17.
Blood ; 117(24): 6552-61, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21511956

RESUMEN

In the bone marrow (BM), stromal cells constitute a supportive tissue indispensable for the generation of pro-B/pre-BI, pre-BII, and immature B lymphocytes. IL-7-producing stromal cells constitute a cellular niche for pro-B/pre-BI cells, but no specific stromal cell microenvironment was identified for pre-BII cells expressing a functional pre-B cell receptor (pre-BCR). However expression of the pre-BCR represents a crucial checkpoint during B-cell development. We recently demonstrated that the stromal cell derived-galectin1 (GAL1) is a ligand for the pre-BCR, involved in the proliferation and differentiation of normal mouse pre-BII cells. Here we show that nonhematopoietic osteoblasts and reticular cells in the BM express GAL1. We observed that pre-BII cells, unlike the other B-cell subsets, were specifically localized in close contact with GAL1(+) reticular cells. We also determined that IL-7(+) and GAL1(+) cells represent 2 distinct mesenchymal populations with different BM localization. These results demonstrate the existence of a pre-BII specific stromal cell niche and indicate that early B cells move from IL-7(+) to GAL1(+) supportive BM niches during their development.


Asunto(s)
Médula Ósea , Galectina 1/metabolismo , Células Precursoras de Linfocitos B/fisiología , Nicho de Células Madre/fisiología , Células del Estroma/fisiología , Animales , Médula Ósea/metabolismo , Médula Ósea/fisiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Diferenciación Celular/inmunología , Células Cultivadas , Proteínas Fluorescentes Verdes/genética , Interleucina-7/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Nicho de Células Madre/citología , Nicho de Células Madre/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
18.
iScience ; 26(6): 106910, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37378323

RESUMEN

Severe acute malnutrition (SAM) is a multifactorial disease affecting millions of children worldwide. It is associated with changes in intestinal physiology, microbiota, and mucosal immunity, emphasizing the need for multidisciplinary studies to unravel its full pathogenesis. We established an experimental model in which weanling mice fed a high-deficiency diet mimic key anthropometric and physiological features of SAM in children. This diet alters the intestinal microbiota (less segmented filamentous bacteria, spatial proximity to epithelium), metabolism (decreased butyrate), and immune cell populations (depletion of LysoDC in Peyer's patches and intestinal Th17 cells). A nutritional intervention leads to a fast zoometric and intestinal physiology recovery but to an incomplete restoration of the intestinal microbiota, metabolism, and immune system. Altogether, we provide a preclinical model of SAM and have identified key markers to target with future interventions during the education of the immune system to improve SAM whole defects.

19.
Nat Commun ; 14(1): 4290, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37463962

RESUMEN

Endo-lysosomes transport along microtubules and clustering in the perinuclear area are two necessary steps for microbes to activate specialized phagocyte functions. We report that RUN and FYVE domain-containing protein 3 (RUFY3) exists as two alternative isoforms distinguishable by the presence of a C-terminal FYVE domain and by their affinity for phosphatidylinositol 3-phosphate on endosomal membranes. The FYVE domain-bearing isoform (iRUFY3) is preferentially expressed in primary immune cells and up-regulated upon activation by microbes and Interferons. iRUFY3 is necessary for ARL8b + /LAMP1+ endo-lysosomes positioning in the pericentriolar organelles cloud of LPS-activated macrophages. We show that iRUFY3 controls macrophages migration, MHC II presentation and responses to Interferon-γ, while being important for intracellular Salmonella replication. Specific inactivation of rufy3 in phagocytes leads to aggravated pathologies in mouse upon LPS injection or bacterial pneumonia. This study highlights the role of iRUFY3 in controlling endo-lysosomal dynamics, which contributes to phagocyte activation and immune response regulation.


Asunto(s)
Presentación de Antígeno , Lipopolisacáridos , Animales , Ratones , Endosomas/metabolismo , Lipopolisacáridos/metabolismo , Lisosomas/metabolismo , Fagocitos
20.
Mol Microbiol ; 80(3): 628-40, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21362067

RESUMEN

The oxidative burst produced by the NADPH oxidase (Phox) is an essential weapon used by host cells to eradicate engulfed pathogens. In Salmonella typhimurium, oxidative stress resistance has been previously proposed to be mediated by the pathogenicity island 2 type III secretion system (T3SS-2), periplasmic superoxide dismutases and cytoplasmic catalases/peroxidases. Here, we fused an OxyR-dependent promoter to the gfp to build the ahpC-gfp transcriptional fusion. This reporter was used to monitor hydrogen peroxide levels as sensed by Salmonella during the course of an infection. We showed that the expression of this fusion was under the exclusive control of reactive oxygen species produced by the host. The ahpC-gfp expression was noticeably modified in the absence of bacterial periplasmic superoxide dismutases or cytoplasmic catalases/peroxidases. Surprisingly, inactivation of the T3SS-2 had no effect on the ahpC-gfp expression. All together, these results led to a model in which Salmonella resistance relies on its arsenal of detoxifying enzymes to cope with Phox-mediated oxidative stress.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Macrófagos/microbiología , Especies Reactivas de Oxígeno/metabolismo , Estallido Respiratorio , Salmonella typhimurium/efectos de los fármacos , Animales , Fusión Artificial Génica , Células Cultivadas , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inactivación Metabólica , Ratones , Ratones Endogámicos C57BL , Viabilidad Microbiana/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/microbiología , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología , Salmonella typhimurium/fisiología , Bazo/microbiología , Superóxidos/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda