Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell ; 184(14): 3774-3793.e25, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34115982

RESUMEN

Cytomegaloviruses (CMVs) have co-evolved with their mammalian hosts for millions of years, leading to remarkable host specificity and high infection prevalence. Macrophages, which already populate barrier tissues in the embryo, are the predominant immune cells at potential CMV entry sites. Here we show that, upon CMV infection, macrophages undergo a morphological, immunophenotypic, and metabolic transformation process with features of stemness, altered migration, enhanced invasiveness, and provision of the cell cycle machinery for viral proliferation. This complex process depends on Wnt signaling and the transcription factor ZEB1. In pulmonary infection, mouse CMV primarily targets and reprograms alveolar macrophages, which alters lung physiology and facilitates primary CMV and secondary bacterial infection by attenuating the inflammatory response. Thus, CMV profoundly perturbs macrophage identity beyond established limits of plasticity and rewires specific differentiation processes, allowing viral spread and impairing innate tissue immunity.


Asunto(s)
Citomegalovirus/fisiología , Macrófagos Alveolares/virología , Animales , Presentación de Antígeno , Efecto Espectador , Ciclo Celular , Línea Celular Transformada , Reprogramación Celular , Citomegalovirus/patogenicidad , Citomegalovirus/ultraestructura , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Proteínas Fluorescentes Verdes/metabolismo , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/ultraestructura , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Células Madre/patología , Replicación Viral/fisiología , Vía de Señalización Wnt
2.
Immunity ; 50(6): 1482-1497.e7, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31201094

RESUMEN

The skin comprises tissue macrophages as the most abundant resident immune cell type. Their diverse tasks including resistance against invading pathogens, attraction of bypassing immune cells from vessels, and tissue repair require dynamic specification. Here, we delineated the postnatal development of dermal macrophages and their differentiation into subsets by adapting single-cell transcriptomics, fate mapping, and imaging. Thereby we identified a phenotypically and transcriptionally distinct subset of prenatally seeded dermal macrophages that self-maintained with very low postnatal exchange by hematopoietic stem cells. These macrophages specifically interacted with sensory nerves and surveilled and trimmed the myelin sheath. Overall, resident dermal macrophages contributed to axon sprouting after mechanical injury. In summary, our data show long-lasting functional specification of macrophages in the dermis that is driven by stepwise adaptation to guiding structures and ensures codevelopment of ontogenetically distinct cells within the same compartment.


Asunto(s)
Diferenciación Celular/inmunología , Vigilancia Inmunológica , Macrófagos/inmunología , Regeneración Nerviosa , Piel/inmunología , Piel/inervación , Animales , Animales Recién Nacidos , Biomarcadores , Receptor 1 de Quimiocinas CX3C/metabolismo , Dermis/citología , Dermis/inmunología , Dermis/metabolismo , Inmunofenotipificación , Macrófagos/metabolismo , Ratones , Piel/citología
3.
J Immunol ; 203(12): 3416-3426, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31732532

RESUMEN

IFN-ß essentially modulates the host response against mucocutaneous colonizers and potential pathogens, such as group B Streptococcus (GBS). It has been reported that the dominant signaling cascade driving IFN-ß in macrophages (MΦ) in streptococcal infection is the cGAS-STING pathway, whereas conventional dendritic cells (DC) exploit endosomal recognition by intracellular TLRs. In this study, we revisited this issue by precisely monitoring the phenotypic dynamics in mixed mouse MΦ/DC cultures with GM-CSF, which requires snapshot definition of cellular identities. We identified four mononuclear phagocyte populations, of which two were transcriptionally and morphologically distinct MΦ-DC-like subsets, and two were transitional types. Notably, GBS induced a TLR7-dependent IFN-ß signal only in MΦ-like but not in DC-like cells. IFN-ß induction did not require live bacteria (i.e., the formation of cytolytic toxins), which are essential for IFN-ß induction via cGAS-STING. In contrast to IFN-ß, GBS induced TNF-α independently of TLR7. Subsequent to the interaction with streptococci, MΦ changed their immunophenotype and gained some typical DC markers and DC-like morphology. In summary, we identify IFN-ß formation as part of the antistreptococcal repertoire of GM-CSF differentiated MΦ in vitro and in vivo and delineate their plasticity.


Asunto(s)
Interferón beta/biosíntesis , Macrófagos/inmunología , Macrófagos/metabolismo , Streptococcus/inmunología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunofenotipificación , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Modelos Biológicos , Fagocitosis , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/microbiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/microbiología
4.
Methods Mol Biol ; 2713: 347-361, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37639135

RESUMEN

Macrophage identity, as defined by epigenetic, transcriptional, proteomic, and functional programs, is greatly impacted by cues originating from the microenvironment. As a consequence, immunophenotyping based on surface marker expression is established and reliable in homeostatic conditions, whereas environmental challenges, in particular infections, severely hamper the determination of identity states. This has become more evident with recent discoveries that macrophage-inherent plasticity may go beyond limits of lineage-defining immunophenotypes. Therefore, transgenic fate mapping tools, such as the phage-derived loxP-cre-system, are essential for the analysis of macrophage adaptation in the tissue under extreme environmental conditions, for example, upon encounter with pathogens. In this chapter, we describe an advanced application of the loxP-cre-system during infection. Here, the host encodes a cell type-specific cre-recombinase, while the pathogen harbors a STOP-floxed fluorescent reporter gene. As an instructive example for the versatility of the system, we demonstrate that alveolar macrophages are predominantly targeted after respiratory tract infection with mouse cytomegalovirus (MCMV). Combined host-pathogen fate mapping not only enables to distinguish between infected and non-infected (bystander) macrophages but also spurs exploration of phenotypic adaptation and tracing of cellular localization in the context of MCMV infection. Moreover, we provide a gating strategy for resolving the diversity of pulmonary immune cell populations.


Asunto(s)
Macrófagos Alveolares , Virosis , Animales , Ratones , Proteómica , Macrófagos , Pulmón
5.
Nat Commun ; 14(1): 2721, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37169749

RESUMEN

While the precise processes underlying a sex bias in the development of central nervous system (CNS) disorders are unknown, there is growing evidence that an early life immune activation can contribute to the disease pathogenesis. When we mimicked an early systemic viral infection or applied murine cytomegalovirus (MCMV) systemically in neonatal female and male mice, only male adolescent mice presented behavioral deficits, including reduced social behavior and cognition. This was paralleled by an increased amount of infiltrating T cells in the brain parenchyma, enhanced interferon-γ (IFNγ) signaling, and epigenetic reprogramming of microglial cells. These microglial cells showed increased phagocytic activity, which resulted in abnormal loss of excitatory synapses within the hippocampal brain region. None of these alterations were seen in female adolescent mice. Our findings underscore the early postnatal period's susceptibility to cause sex-dependent long-term CNS deficiencies following infections.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Microglía , Animales , Femenino , Masculino , Ratones , Microglía/patología , Encéfalo , Enfermedades del Sistema Nervioso Central/patología , Interferón gamma/genética , Epigénesis Genética
6.
Sci Immunol ; 8(86): eadg3517, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37566679

RESUMEN

The skin needs to balance tolerance of colonizing microflora with rapid detection of potential pathogens. Flexible response mechanisms would seem most suitable to accommodate the dynamic challenges of effective antimicrobial defense and restoration of tissue homeostasis. Here, we dissected macrophage-intrinsic mechanisms and microenvironmental cues that tune macrophage signaling in localized skin infection with the colonizing and opportunistic pathogen Staphylococcus aureus. Early in skin infection, the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) produced by γδ T cells and hypoxic conditions within the dermal microenvironment diverted macrophages away from a homeostatic M-CSF- and hypoxia-inducible factor 1α (HIF-1α)-dependent program. This allowed macrophages to be metabolically rewired for maximal inflammatory activity, which requires expression of Irg1 and generation of itaconate, but not HIF-1α. This multifactorial macrophage rewiring program was required for both the timely clearance of bacteria and for the provision of local immune memory. These findings indicate that immunometabolic conditioning allows dermal macrophages to cycle between antimicrobial activity and protection against secondary infections.


Asunto(s)
Macrófagos , Infecciones Cutáneas Estafilocócicas , Humanos , Citocinas/metabolismo , Transducción de Señal , Infecciones Cutáneas Estafilocócicas/metabolismo
7.
Front Immunol ; 11: 793, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32477336

RESUMEN

Starting at birth, newborn infants are exposed to numerous microorganisms. Adaptation of the innate immune system to them is a delicate process, with potentially advantageous and harmful implications for health development. Cytomegaloviruses (CMVs) are highly adapted to their specific mammalian hosts, with which they share millions of years of co-evolution. Throughout the history of mankind, human CMV has infected most infants in the first months of life without overt implications for health. Thus, CMV infections are intertwined with normal immune development. Nonetheless, CMV has retained substantial pathogenicity following infection in utero or in situations of immunosuppression, leading to pathology in virtually any organ and particularly the central nervous system (CNS). CMVs enter the host through mucosal interfaces of the gastrointestinal and respiratory tract, where macrophages (MACs) are the most abundant immune cell type. Tissue MACs and their potential progenitors, monocytes, are established target cells of CMVs. Recently, several discoveries have revolutionized our understanding on the pre- and postnatal development and site-specific adaptation of tissue MACs. In this review, we explore experimental evidences and concepts on how CMV infections may impact on MAC development and activation as part of host-virus co-adaptation.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Inmunidad Innata , Inmunidad Mucosa , Macrófagos/inmunología , Animales , Infecciones por Citomegalovirus/virología , Adaptación al Huésped/inmunología , Humanos , Inmunomodulación , Lactante , Recién Nacido , Ratones , Monocitos/inmunología
8.
Acta Neuropathol Commun ; 2: 169, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25519173

RESUMEN

INTRODUCTION: Cerebral small vessel disease (cSVD) is one of the most prevalent neurological disorders. The progressive remodeling of brain microvessels due to arterial hypertension or other vascular risk factors causes subtle, but constant cognitive decline through to manifest dementia and substantially increases the risk for stroke. Preliminary evidence suggests the contribution of the immune system to disease initiation and progression, but a more detailed understanding is impaired by the unavailability of appropriate animal models. Here, we introduce the spontaneously hypertensive rat (SHR) as a model for early onset cSVD and unveiled substantial immune changes in conjunction with brain abnormalities that resemble clinical findings. RESULTS: In contrast to age-matched normotensive Wistar Kyoto (WKY) rats, male SHR exhibited non-spatial memory deficits. Magnetic resonance imaging showed brain atrophy and a reduction of white matter volumes in SHR. Histological analyses confirmed white matter demyelination and unveiled a circumscribed blood brain barrier dysfunction in conjunction with micro- and macrogliosis in deep cortical regions. Flow cytometry and histological analyses further revealed substantial disparities in cerebral CD45high leukocyte counts and distribution patterns between SHR and WKY. SHR showed lower counts of T cells in the choroid plexus and meningeal spaces as well as decreased interleukin-10 levels in the cerebrospinal fluid. On the other hand, both T and NK cells were significantly augmented in the SHR brain microvasculature. CONCLUSIONS: Our results indicate that SHR share behavioral and neuropathological characteristics with human cSVD patients and further undergird the relevance of immune responses for the initiation and progression of cSVD.


Asunto(s)
Encéfalo/patología , Enfermedades de los Pequeños Vasos Cerebrales/fisiopatología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Neuroinmunomodulación/fisiología , Sustancia Blanca/patología , Animales , Atrofia , Barrera Hematoencefálica/fisiopatología , Encéfalo/fisiopatología , Enfermedades de los Pequeños Vasos Cerebrales/patología , Enfermedades de los Pequeños Vasos Cerebrales/psicología , Trastornos del Conocimiento/patología , Gliosis/inmunología , Gliosis/patología , Interleucina-10/líquido cefalorraquídeo , Células Asesinas Naturales/patología , Células Asesinas Naturales/fisiología , Antígenos Comunes de Leucocito/metabolismo , Leucocitos/patología , Leucocitos/fisiología , Masculino , Trastornos de la Memoria/inmunología , Trastornos de la Memoria/patología , Tamaño de los Órganos , Distribución Aleatoria , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Linfocitos T/patología , Linfocitos T/fisiología , Sustancia Blanca/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA