Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Ann Surg ; 278(6): e1289-e1298, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37325925

RESUMEN

OBJECTIVE: To characterize the role of neutrophil extracellular traps (NETs) in heterotopic ossification (HO) formation and progression and to use mechanical and pharmacological methods to decrease NETosis and mitigate HO formation. BACKGROUND: Traumatic HO is the aberrant osteochondral differentiation of mesenchymal progenitor cells after traumatic injury, burns, or surgery. While the innate immune response has been shown to be necessary for HO formation, the specific immune cell phenotype and function remain unknown. Neutrophils, one of the earliest immune cells to respond after HO-inducing injuries, can extrude DNA, forming highly inflammatory NETs. We hypothesized that neutrophils and NETs would be diagnostic biomarkers and therapeutic targets for the detection and mitigation of HO. METHODS: C57BL6J mice underwent burn/tenotomy (a well-established mouse model of HO) or a non-HO-forming sham injury. These mice were either (1) ambulated ad libitum, (2) ambulated ad libitum with daily intraperitoneal hydroxychloroquine, ODN-2088 (both known to affect NETosis pathways), or control injections, or (3) had the injured hind limb immobilized. Single-cell analysis was performed to analyze neutrophils, NETosis, and downstream signaling after the HO-forming injury. Immunofluorescence microscopy was used to visualize NETosis at the HO site and neutrophils were identified using flow cytometry. Serum and cell lysates from HO sites were analyzed using enzyme-linked immunosorbent assay for myeloperoxidase-DNA and ELA2-DNA complexes to identify NETosis. Micro-computerized tomography was performed on all groups to analyze the HO volume. RESULTS: Molecular and transcriptional analyses revealed the presence of NETs within the HO injury site, which peaked in the early phases after injury. These NETs were highly restricted to the HO site, with gene signatures derived from both in vitro NET induction and clinical neutrophil characterizations showing a high degree of NET "priming" at the site of injury, but not in neutrophils in the blood or bone marrow. Cell-cell communication analyses revealed that this localized NET formation coincided with high levels of toll-like receptor signaling specific to neutrophils at the injury site. Reducing the overall neutrophil abundance within the injury site, either pharmacologically through treatment with hydroxychloroquine, the toll-like receptor 9 inhibitor OPN-2088, or mechanical treatment with limb offloading, results in the mitigation of HO formation. CONCLUSIONS: These data provide a further understanding of the ability of neutrophils to form NETs at the injury site, clarify the role of neutrophils in HO, and identify potential diagnostic and therapeutic targets for HO mitigation.


Asunto(s)
Trampas Extracelulares , Neutrófilos , Animales , Ratones , Neutrófilos/metabolismo , Hidroxicloroquina/metabolismo , Trampas Extracelulares/metabolismo , Inmunidad Innata , ADN/metabolismo
2.
J Immunol ; 204(8): 2203-2215, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32161098

RESUMEN

Myeloid cells are critical to the development of fibrosis following muscle injury; however, the mechanism of their role in fibrosis formation remains unclear. In this study, we demonstrate that myeloid cell-derived TGF-ß1 signaling is increased in a profibrotic ischemia reperfusion and cardiotoxin muscle injury model. We found that myeloid-specific deletion of Tgfb1 abrogates the fibrotic response in this injury model and reduces fibro/adipogenic progenitor cell proliferation while simultaneously enhancing muscle regeneration, which is abrogated by adaptive transfer of normal macrophages. Similarly, a murine TGFBRII-Fc ligand trap administered after injury significantly reduced muscle fibrosis and improved muscle regeneration. This study ultimately demonstrates that infiltrating myeloid cell TGF-ß1 is responsible for the development of traumatic muscle fibrosis, and its blockade offers a promising therapeutic target for preventing muscle fibrosis after ischemic injury.


Asunto(s)
Fibrosis/inmunología , Fibrosis/patología , Macrófagos/inmunología , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Células Mieloides/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Animales , Cardiotoxinas , Fibrosis/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/patología , Fenotipo , Daño por Reperfusión/inducido químicamente , Daño por Reperfusión/complicaciones , Daño por Reperfusión/inmunología
3.
Am J Pathol ; 190(9): 1909-1920, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32533926

RESUMEN

Perivascular mural cells surround capillaries and microvessels and have diverse regenerative or fibrotic functions after tissue injury. Subsynovial fibrosis is a well-known pathologic feature of osteoarthritis, yet transgenic animals for use in visualizing perivascular cell contribution to fibrosis during arthritic changes have not been developed. Here, inducible Pdgfra-CreERT2 reporter mice were subjected to joint-destabilization surgery to induce arthritic changes, and cell lineage was traced over an 8-week period with a focus on the joint-associated fat pad. Results showed that, at baseline, inducible Pdgfra reporter activity highlighted adventitial and, to a lesser extent, pericytic cells within the infrapatellar fat pad. Joint-destabilization surgery was associated with marked fibrosis of the infrapatellar fat pad, accompanied by an expansion of perivascular Pdgfra-expressing cellular descendants, many of which adopted α-smooth muscle actin expression. Gene expression analysis of microdissected infrapatellar fat pad confirmed enrichment in membrane-bound green fluorescent protein/Pdgfra-expressing cells, along with a gene signature that corresponded with injury-associated fibro-adipogenic progenitors. Our results highlight dynamic changes in joint-associated perivascular fibro-adipogenic progenitors during osteoarthritis.


Asunto(s)
Adipocitos/patología , Fibroblastos/patología , Osteoartritis/patología , Tejido Adiposo/patología , Animales , Linaje de la Célula , Articulación de la Rodilla/patología , Ratones , Ratones Transgénicos , Células Madre
4.
FASEB J ; 34(12): 15753-15770, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33089917

RESUMEN

Ischemia reperfusion (IR) injury results in devastating skeletal muscle fibrosis. Here, we recapitulate this injury with a mouse model of hindlimb IR injury which leads to skeletal muscle fibrosis. Injury resulted in extensive immune infiltration with robust neutrophil extracellular trap (NET) formation in the skeletal muscle, however, direct targeting of NETs via the peptidylarginine deiminase 4 (PAD4) mechanism was insufficient to reduce muscle fibrosis. Circulating levels of IL-10 and TNFα were significantly elevated post injury, indicating toll-like receptor (TLR) signaling may be involved in muscle injury. Administration of hydroxychloroquine (HCQ), a small molecule inhibitor of TLR7/8/9, following injury reduced NET formation, IL-10, and TNFα levels and ultimately mitigated muscle fibrosis and improved myofiber regeneration following IR injury. HCQ treatment decreased fibroadipogenic progenitor cell proliferation and partially inhibited ERK1/2 phosphorylation in the injured tissue, suggesting it may act through a combination of TLR7/8/9 and ERK signaling mechanisms. We demonstrate that treatment with FDA-approved HCQ leads to decreased muscle fibrosis and increased myofiber regeneration following IR injury, suggesting short-term HCQ treatment may be a viable treatment to prevent muscle fibrosis in ischemia reperfusion and traumatic extremity injury.


Asunto(s)
Trampas Extracelulares/metabolismo , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Neutrófilos/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal/fisiología , Receptores Toll-Like/metabolismo , Animales , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Fibrosis/metabolismo , Interleucina-10/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Arginina Deiminasa Proteína-Tipo 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Immunol ; 200(3): 966-973, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29288202

RESUMEN

GM-CSF has been portrayed as a critical cytokine in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and, ostensibly, in multiple sclerosis. C57BL/6 mice deficient in GM-CSF are resistant to EAE induced by immunization with myelin oligodendrocyte glycoprotein (MOG)35-55 The mechanism of action of GM-CSF in EAE is poorly understood. In this study, we show that GM-CSF augments the accumulation of MOG35-55-specific T cells in the skin draining lymph nodes of primed mice, but it is not required for the development of encephalitogenic T cells. Abrogation of GM-CSF receptor signaling in adoptive transfer recipients of MOG35-55-specific T cells did not alter the incidence of EAE or the trajectory of its initial clinical course, but it limited the extent of chronic CNS tissue damage and neurologic disability. The attenuated clinical course was associated with a relative dearth of MOG35-55-specific T cells, myeloid dendritic cells, and neutrophils, as well as an abundance of B cells, within CNS infiltrates. Our data indicate that GM-CSF drives chronic tissue damage and disability in EAE via pleiotropic pathways, but it is dispensable during early lesion formation and the onset of neurologic deficits.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Sistema Nervioso Central/citología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/trasplante , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Pulmón/patología , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Neutrófilos/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
6.
Clin Immunol ; 189: 29-33, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-27664933

RESUMEN

Eotaxins are C-C motif chemokines first identified as potent eosinophil chemoattractants. They facilitate eosinophil recruitment to sites of inflammation in response to parasitic infections as well as allergic and autoimmune diseases such as asthma, atopic dermatitis, and inflammatory bowel disease. The eotaxin family currently includes three members: eotaxin-1 (CCL11), eotaxin-2 (CCL24), and eotaxin-3 (CCL26). Despite having only ~30% sequence homology to one another, each was identified based on its ability to bind the chemokine receptor, CCR3. Beyond their role in innate immunity, recent studies have shown that CCL11 and related molecules may directly contribute to degenerative processes in the central nervous system (CNS). CCL11 levels increase in the plasma and cerebrospinal fluid of both mice and humans as part of normal aging. In mice, these increases are associated with declining neurogenesis and impaired cognition and memory. In humans, elevated plasma levels of CCL11 have been observed in Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's disease, and secondary progressive multiple sclerosis when compared to age-matched, healthy controls. Since CCL11 is capable of crossing the blood-brain barrier of normal mice, it is plausible that eotaxins generated in the periphery may exert physiological and pathological actions in the CNS. Here, we briefly review known functions of eotaxin family members during innate immunity, and then focus on whether and how these molecules might participate in the progression of neurodegenerative diseases.


Asunto(s)
Quimiocina CCL11/inmunología , Quimiocina CCL24/inmunología , Quimiocina CCL26/inmunología , Inmunidad Innata/inmunología , Enfermedades Neurodegenerativas/inmunología , Envejecimiento/inmunología , Animales , Quimiocina CCL11/sangre , Quimiocina CCL11/líquido cefalorraquídeo , Quimiocina CCL24/sangre , Quimiocina CCL24/líquido cefalorraquídeo , Quimiocina CCL26/sangre , Quimiocina CCL26/líquido cefalorraquídeo , Humanos , Enfermedades Neurodegenerativas/sangre , Enfermedades Neurodegenerativas/líquido cefalorraquídeo , Receptores CCR3/inmunología , Receptores CCR3/metabolismo
7.
J Neuroinflammation ; 15(1): 208, 2018 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-30012158

RESUMEN

BACKGROUND: Myelin oligodendrocyte glycoprotein (MOG)-reactive T-helper (Th)1 cells induce conventional experimental autoimmune encephalomyelitis (cEAE), characterized by ascending paralysis and monocyte-predominant spinal cord infiltrates, in C57BL/6 wildtype (WT) hosts. The same T cells induce an atypical form of EAE (aEAE), characterized by ataxia and neutrophil-predominant brainstem infiltrates, in syngeneic IFNγ receptor (IFNγR)-deficient hosts. Production of ELR+ CXC chemokines within the CNS is required for the development of aEAE, but not cEAE. The cellular source(s) and localization of ELR+ CXC chemokines in the CNS and the IFNγ-dependent pathways that regulate their production remain to be elucidated. METHODS: The spatial distribution of inflammatory lesions and CNS expression of the ELR+ CXC chemokines, CXCL1 and CXCL2, were determined via immunohistochemistry and/or in situ hybridization. Levels of CXCL1 and CXCL2, and their cognate receptor CXCR2, were measured in/on leukocyte subsets by flow cytometric and quantitative PCR (qPCR) analysis. Bone marrow neutrophils and macrophages were cultured with inflammatory stimuli in vitro prior to measurement of CXCL2 and CXCR2 by qPCR or flow cytometry. RESULTS: CNS-infiltrating neutrophils and monocytes, and resident microglia, are a prominent source of CXCL2 in the brainstem of IFNγRKO adoptive transfer recipients during aEAE. In WT transfer recipients, IFNγ directly suppresses CXCL2 transcription in microglia and myeloid cells, and CXCR2 transcription in CNS-infiltrating neutrophils. Consequently, infiltration of the brainstem parenchyma from the adjacent meninges is blocked during cEAE. CXCL2 directly stimulates its own expression in cultured neutrophils, which is enhanced by IL-1 and suppressed by IFNγ. CONCLUSIONS: We provide evidence for an IFNγ-regulated CXCR2/CXCL2 autocrine/paracrine feedback loop in innate immune cells that determines the location of CNS infiltrates during Th1-mediated EAE. When IFNγ signaling is impaired, myeloid cell production of CXCL2 increases, which promotes brainstem inflammation and results in clinical ataxia. IFNγ, produced within the CNS of WT recipients, suppresses myeloid cell CXCR2 and CXCL2 production, thereby skewing the location of neuroinflammatory infiltrates to the spinal cord and the clinical phenotype to an ascending paralysis. These data reveal a novel mechanism by which IFNγ and CXCL2 interact to direct regional recruitment of leukocytes in the CNS, resulting in distinct clinical presentations.


Asunto(s)
Encéfalo/metabolismo , Quimiocina CXCL2/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Interferón gamma/metabolismo , Transducción de Señal/fisiología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Proteína Ácida Fibrilar de la Glía/metabolismo , Interferón gamma/genética , Interferón gamma/farmacología , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/efectos de los fármacos , Monocitos/patología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Células Mieloides/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/patología , Tejido Parenquimatoso/patología , Fragmentos de Péptidos/toxicidad , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos
8.
J Biol Chem ; 291(6): 3030-42, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26601958

RESUMEN

UBE2W ubiquitinates N termini of proteins rather than internal lysine residues, showing a preference for substrates with intrinsically disordered N termini. The in vivo functions of this intriguing E2, however, remain unknown. We generated Ube2w germ line KO mice that proved to be susceptible to early postnatal lethality without obvious developmental abnormalities. Although the basis of early death is uncertain, several organ systems manifest changes in Ube2w KO mice. Newborn Ube2w KO mice often show altered epidermal maturation with reduced expression of differentiation markers. Mirroring higher UBE2W expression levels in testis and thymus, Ube2w KO mice showed a disproportionate decrease in weight of these two organs (~50%), suggesting a functional role for UBE2W in the immune and male reproductive systems. Indeed, Ube2w KO mice displayed sustained neutrophilia accompanied by increased G-CSF signaling and testicular vacuolation associated with decreased fertility. Proteomic analysis of a vulnerable organ, presymptomatic testis, showed a preferential accumulation of disordered proteins in the absence of UBE2W, consistent with the view that UBE2W preferentially targets disordered polypeptides. These mice further allowed us to establish that UBE2W is ubiquitously expressed as a single isoform localized to the cytoplasm and that the absence of UBE2W does not alter cell viability in response to various stressors. Our results establish that UBE2W is an important, albeit not essential, protein for early postnatal survival and normal functioning of multiple organ systems.


Asunto(s)
Epidermis , Anomalías Cutáneas , Enzimas Ubiquitina-Conjugadoras , Animales , Epidermis/anomalías , Epidermis/enzimología , Epidermis/inmunología , Trastornos Leucocíticos/congénito , Trastornos Leucocíticos/enzimología , Trastornos Leucocíticos/genética , Trastornos Leucocíticos/inmunología , Masculino , Ratones , Ratones Noqueados , Anomalías Cutáneas/enzimología , Anomalías Cutáneas/genética , Anomalías Cutáneas/inmunología , Testículo/enzimología , Testículo/inmunología , Timo/enzimología , Timo/inmunología , Enzimas Ubiquitina-Conjugadoras/deficiencia , Enzimas Ubiquitina-Conjugadoras/inmunología
9.
J Immunol ; 195(6): 2552-9, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26238492

RESUMEN

Multiple sclerosis (MS) is believed to be initiated by myelin-reactive CD4(+) Th cells. IL-12-polarized Th1 cells, IL-23-polarized Th17 cells, and Th17 cells that acquire Th1 characteristics were each implicated in autoimmune pathogenesis. It is debated whether Th cells that can drive the development of demyelinating lesions are phenotypically diverse or arise from a single lineage. In the current study, we assessed the requirement of IL-12 or IL-23 stimulation, as well as Th plasticity, for the differentiation of T cells capable of inducing CNS axon damage. We found that stable murine Th1 and Th17 cells independently transfer experimental autoimmune encephalomyelitis (widely used as an animal model of MS) in the absence of IL-23 and IL-12, respectively. Plastic Th17 cells are particularly potent mediators of demyelination and axonopathy. In parallel studies, we identified MS patients who consistently mount either IFN-γ- or IL-17-skewed responses to myelin basic protein over the course of a year. Brain magnetic resonance imaging revealed that patients with mixed IFN-γ and IL-17 responses have relatively high T1 lesion burden, a measure of permanent axon damage. Our data challenge the dogma that IL-23 and Th17 plasticity are universally required for the development of experimental autoimmune encephalomyelitis. This study definitively demonstrates that autoimmune demyelinating disease can be driven by distinct Th-polarizing factors and effector subsets, underscoring the importance of a customized approach to the pharmaceutical management of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Células TH1/inmunología , Células Th17/inmunología , Traslado Adoptivo , Animales , Autoinmunidad/inmunología , Encéfalo/diagnóstico por imagen , Diferenciación Celular/inmunología , Enfermedades Desmielinizantes/inmunología , Humanos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-17/inmunología , Interleucina-23/inmunología , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Básica de Mielina/inmunología , Nervio Óptico/inmunología , Nervio Óptico/patología , Radiografía , Células TH1/citología , Células TH1/trasplante , Células Th17/citología , Células Th17/trasplante
10.
J Neuroinflammation ; 13(1): 196, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27562117

RESUMEN

BACKGROUND: Alphaviruses can cause fatal encephalitis in humans. Natural infections occur via the bite of infected mosquitos, but aerosol transmissibility makes some of these viruses potential bioterrorism agents. Central nervous system (CNS) host responses contribute to alphavirus pathogenesis in experimental models and are logical therapeutic targets. We investigated whether reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) activity within the CNS contributes to fatal alphavirus encephalitis in mice. METHODS: Infected animals were treated systemically with the angiotensin receptor-blocking drug, telmisartan, given its ability to cross the blood-brain barrier, selectively block type-1 angiotensin receptors (AT1R), and inhibit Nox-derived ROS production in vascular smooth muscle and other extraneural tissues. Clinical, virological, biochemical, and histopathological outcomes were followed over time. RESULTS: The importance of the angiotensin II (Ang II)/AT1R axis in disease pathogenesis was confirmed by demonstrating increased Ang II levels in the CNS following infection, enhanced disease survival when CNS Ang II production was suppressed, increased AT1R expression on microglia and tissue-infiltrating myeloid cells, and enhanced disease survival in AT1R-deficient mice compared to wild-type (WT) controls. Systemic administration of telmisartan protected WT mice from lethal encephalitis caused by two different alphaviruses in a dose-dependent manner without altering virus replication or exerting any anti-inflammatory effects in the CNS. Infection triggered up-regulation of multiple Nox subunits in the CNS, while drug treatment inhibited local Nox activity, ROS production, and oxidative neuronal damage. Telmisartan proved ineffective in Nox-deficient mice, demonstrating that this enzyme is its main target in this experimental setting. CONCLUSIONS: Nox-derived ROS, likely arising from CNS myeloid cells triggered by AT1R signaling, are pathogenic during fatal alphavirus encephalitis in mice. Systemically administered telmisartan at non-hypotensive doses targets Nox activity in the CNS to exert a neuroprotective effect. Disruption of this pathway may have broader implications for the treatment of related infections as well as for other CNS diseases driven by oxidative injury.


Asunto(s)
Sistema Nervioso Central/patología , Encefalomielitis Equina/patología , Células Mieloides/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/fisiología , ATPasas Asociadas con Actividades Celulares Diversas , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Bencimidazoles/farmacología , Benzoatos/farmacología , Receptor 1 de Quimiocinas CX3C , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/virología , ADN Helicasas/genética , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Equina/tratamiento farmacológico , Encefalomielitis Equina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/clasificación , Células Mieloides/ultraestructura , Células Mieloides/virología , Neuronas/patología , Neuronas/ultraestructura , Consumo de Oxígeno/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Telmisartán
11.
J Immunol ; 193(2): 564-70, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24928987

RESUMEN

The adoptive transfer of myelin-reactive T cells into wild-type hosts results in spinal cord inflammation and ascending paralysis, referred to as conventional experimental autoimmune encephalomyelitis (EAE), as opposed to brainstem inflammation and ataxia, which characterize disease in IFN-γRKO hosts (atypical EAE). In this article, we show that atypical EAE correlates with preferential upregulation of CXCL2 in the brainstem, and is driven by CXCR2-dependent recruitment of neutrophils. In contrast, conventional EAE is associated with upregulation of CCL2 in the spinal cord, and is driven by recruitment of monocytes via a partially CCR2-dependent pathway. This study illustrates how regional differences in chemokine expression within a target organ shape the spatial pattern and composition of autoimmune infiltrates, leading to disparate clinical outcomes.


Asunto(s)
Sistema Nervioso Central/inmunología , Quimiocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/inmunología , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígenos de Diferenciación de Linfocitos T/metabolismo , Tronco Encefálico/inmunología , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Quimiocinas/biosíntesis , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Citometría de Flujo , Inmunofenotipificación , Inflamación/genética , Inflamación/metabolismo , Interleucina-12/inmunología , Interleucina-12/metabolismo , Interleucina-17/inmunología , Interleucina-17/metabolismo , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Monocitos/inmunología , Monocitos/metabolismo , Glicoproteína Mielina-Oligodendrócito , Neutrófilos/inmunología , Neutrófilos/metabolismo , Fragmentos de Péptidos , Receptores CCR2/deficiencia , Receptores CCR2/genética , Receptores CCR2/inmunología , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Receptores de Interleucina-8B/inmunología , Receptores de Interleucina-8B/metabolismo , Receptor de Interferón gamma
12.
Cytokine ; 74(2): 228-36, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25691330

RESUMEN

A series of controlled clinical trials have shown that exogenous interferon-beta (IFN-ß) benefits patients with relapsing-remitting multiple sclerosis (RRMS) by reducing relapse rate, disability progression, and the formation of new brain and spinal cord lesions on magnetic resonance imaging (MRI) scans. Unfortunately, however, the effectiveness of IFN-ß is limited in this setting by the occurrence of treatment non-responsiveness in nearly 25% of patients. Furthermore, clinicians who care for RRMS patients remain unable to accurately identify IFN-ß non-responders prior to the initiation of therapy, causing delays in the use of alternative treatments and sometimes requiring that patients turn to medications with more significant side effects to control their disease. Progress has been made toward understanding how both endogenous and exogenous IFN-ß act to slow RRMS as well as the related mouse model, experimental autoimmune encephalomyelitis (EAE). Most studies point to its inhibitory actions on circulating immune cells as being important for suppressing both disorders, but multiple potential target cells and inflammatory pathways have been implicated and those essential to confer its benefits remain undefined. This review focuses on the role of both endogenous and exogenous IFN-ß in RRMS, paying particular attention to the issue of why certain individuals appear refractory to its disease-modifying effects. A continued goal in this field remains the identification of a convenient biomarker that accurately predicts IFN-ß treatment non-responsiveness in individual RRMS patients. Development of such an assay will allow clinicians to customize therapy for patients with this complex disorder.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Interferón beta , Esclerosis Múltiple , Animales , Ensayos Clínicos como Asunto , Encefalomielitis Autoinmune Experimental/diagnóstico por imagen , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Interferón beta/inmunología , Interferón beta/uso terapéutico , Ratones , Ratones Noqueados , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Radiografía
13.
Glia ; 62(9): 1452-62, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24829092

RESUMEN

Lymphoid chemokines are crucial for the development and maintenance of lymphoid organs, but their ectopic expression in non-lymphoid tissues is implicated in both local response to infection and chronic organ-specific autoimmunity. Production of one such chemokine, C-X-C motif ligand 13 (CXCL13), within the central nervous system (CNS) has been linked to the pathogenesis of multiple sclerosis (MS), although little is known about factors controlling its expression in different neural cell types and across a range of disease states. We provoked acute neuroinflammation in experimental animals without causing any associated demyelination using neuroadapted Sindbis virus (NSV) to better understand the sources and regulators of this chemokine in the CNS. We found that mice genetically deficient in the transcription factor, interferon (IFN) regulatory factor-7 (IRF7), made significantly higher CXCL13 protein levels in the CNS compared with wild-type (WT) controls. Microglia proved to be the main producer of CXCL13 in the brain during infection of both WT and IRF7(-/-) mice, and primary microglia cultured in vitro generated CXCL13 following stimulation with either virus particles or synthetic Toll-like receptor (TLR) ligands. Microglia cultured from IRF7(-/-) mice selectively overproduced CXCL13, and manipulation of extracellular type-I IFN levels demonstrated the existence of a negative feedback loop whereby type-I IFN receptor signaling specifically suppressed microglial CXCL13 release. Since IFN-ß is used to treat patients with relapsing-remitting MS and yet acts through unknown mechanisms, we speculate that suppressed lymphoid chemokine production by microglia could contribute to its therapeutic effects.


Asunto(s)
Encéfalo/inmunología , Quimiocina CXCL13/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Microglía/metabolismo , Infecciones por Alphavirus/inmunología , Animales , Células Cultivadas , Quimiocina CXCL13/genética , Modelos Animales de Enfermedad , Encefalitis Viral/inmunología , Encefalomielitis/inmunología , Factor 7 Regulador del Interferón/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación/fisiología , Virus Sindbis , Receptores Toll-Like/metabolismo
14.
J Immunol ; 189(6): 3043-53, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22888137

RESUMEN

The CD40 gene, an important immune regulatory gene, is also expressed and functional on nonmyeloid-derived cells, many of which are targets for tissue-specific autoimmune diseases, including ß cells in type 1 diabetes, intestinal epithelial cells in Crohn's disease, and thyroid follicular cells in Graves' disease (GD). Whether target tissue CD40 expression plays a role in autoimmune disease etiology has yet to be determined. In this study, we show that target tissue overexpression of CD40 plays a key role in the etiology of autoimmunity. Using a murine model of GD, we demonstrated that thyroidal CD40 overexpression augmented the production of thyroid-specific Abs, resulting in more severe experimental autoimmune GD (EAGD), whereas deletion of thyroidal CD40 suppressed disease. Using transcriptome and immune-pathway analyses, we showed that in both EAGD mouse thyroids and human primary thyrocytes, CD40 mediates this effect by activating downstream cytokines and chemokines, most notably IL-6. To translate these findings into therapy, we blocked IL-6 during EAGD induction in the setting of thyroidal CD40 overexpression and showed decreased levels of thyroid stimulating hormone receptor-stimulating Abs and frequency of disease. We conclude that target tissue overexpression of CD40 plays a key role in the etiology of organ-specific autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Antígenos CD40/genética , Marcación de Gen/métodos , Enfermedad de Graves/genética , Enfermedad de Graves/inmunología , Animales , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/prevención & control , Antígenos CD40/biosíntesis , Antígenos CD40/deficiencia , Células Cultivadas , Modelos Animales de Enfermedad , Enfermedad de Graves/prevención & control , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Cultivo Primario de Células , Quimera por Radiación/inmunología , Receptores de Tirotropina/inmunología , Glándula Tiroides/inmunología , Glándula Tiroides/metabolismo , Glándula Tiroides/patología
15.
JCI Insight ; 9(6)2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38376927

RESUMEN

Radiotherapy induces a type I interferon-mediated (T1IFN-mediated) antitumoral immune response that we hypothesized could be potentiated by a first-in-class ataxia telangiectasia mutated (ATM) inhibitor, leading to enhanced innate immune signaling, T1IFN expression, and sensitization to immunotherapy in pancreatic cancer. We evaluated the effects of AZD1390 or a structurally related compound, AZD0156, on innate immune signaling and found that both inhibitors enhanced radiation-induced T1IFN expression via the POLIII/RIG-I/MAVS pathway. In immunocompetent syngeneic mouse models of pancreatic cancer, ATM inhibitor enhanced radiation-induced antitumoral immune responses and sensitized tumors to anti-PD-L1, producing immunogenic memory and durable tumor control. Therapeutic responses were associated with increased intratumoral CD8+ T cell frequency and effector function. Tumor control was dependent on CD8+ T cells, as therapeutic efficacy was blunted in CD8+ T cell-depleted mice. Adaptive immune responses to combination therapy provided systemic control of contralateral tumors outside of the radiation field. Taken together, we show that a clinical candidate ATM inhibitor enhances radiation-induced T1IFN, leading to both innate and subsequent adaptive antitumoral immune responses and sensitization of otherwise resistant pancreatic cancer to immunotherapy.


Asunto(s)
Ataxia Telangiectasia , Interferón Tipo I , Neoplasias Pancreáticas , Piridinas , Quinolonas , Animales , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Neoplasias Pancreáticas/patología , Inmunidad
16.
J Immunol ; 186(8): 4693-706, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21402899

RESUMEN

IFN-α is known to play a key role in autoimmunity, but the mechanisms are uncertain. Although the induction of autoimmunity by IFN-α is consistent with primarily immunomodulatory effects, the high frequency of nonautoimmune inflammation suggests other mechanisms. We used thyroiditis as a model to dissect these possibilities. IFN-α treatment of cultured thyrocytes increased expression of thyroid differentiation markers, thyroglobulin, thyroid-stimulating hormone receptor, thyroid peroxidase, and sodium iodide transporter. RNAseq analysis demonstrated that pathways of Ag presentation, pattern recognition receptors, and cytokines/chemokines were also stimulated. These changes were associated with markedly increased nonapoptotic thyroid cell death, suggesting direct toxicity. To corroborate these in vitro findings, we created transgenic mice with thyroid-specific overexpression of IFN-α under control of the thyroglobulin promoter. Transgenic mice developed marked inflammatory thyroid destruction associated with immune cell infiltration of thyroid and surrounding tissues leading to profound hypothyroidism, findings consistent with our in vitro results. In addition, transgenic mice thyroids showed upregulation of pathways similar to those observed in cultured thyrocytes. In particular, expression of granzyme B, CXCL10, a subset of the tripartite motif-containing family, and other genes involved in recruitment of bystander cytotoxic immune responses were increased. Pathways associated with apoptosis and autophagy were not induced. Taken together, our data demonstrate that the induction of tissue inflammation and autoimmunity by IFN-α involves direct tissue toxic effects as well as provocation of destructive bystander immune responses.


Asunto(s)
Autoinmunidad/inmunología , Sistema Inmunológico/inmunología , Interferón-alfa/inmunología , Glándula Tiroides/inmunología , Animales , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/metabolismo , Interferón-alfa/genética , Interferón-alfa/farmacología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Ratas , Receptor de Interferón alfa y beta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Glándula Tiroides/citología , Glándula Tiroides/metabolismo , Tiroiditis/genética , Tiroiditis/metabolismo , Factores de Tiempo
17.
Cell Metab ; 35(1): 134-149.e6, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36528023

RESUMEN

Effective therapies are lacking for patients with advanced colorectal cancer (CRC). The CRC tumor microenvironment has elevated metabolic waste products due to altered metabolism and proximity to the microbiota. The role of metabolite waste in tumor development, progression, and treatment resistance is unclear. We generated an autochthonous metastatic mouse model of CRC and used unbiased multi-omic analyses to reveal a robust accumulation of tumoral ammonia. The high ammonia levels induce T cell metabolic reprogramming, increase exhaustion, and decrease proliferation. CRC patients have increased serum ammonia, and the ammonia-related gene signature correlates with altered T cell response, adverse patient outcomes, and lack of response to immune checkpoint blockade. We demonstrate that enhancing ammonia clearance reactivates T cells, decreases tumor growth, and extends survival. Moreover, decreasing tumor-associated ammonia enhances anti-PD-L1 efficacy. These findings indicate that enhancing ammonia detoxification can reactivate T cells, highlighting a new approach to enhance the efficacy of immunotherapies.


Asunto(s)
Amoníaco , Neoplasias Colorrectales , Animales , Ratones , Agotamiento de Células T , Linfocitos T , Neoplasias Colorrectales/patología , Inmunoterapia , Microambiente Tumoral
18.
J Biol Chem ; 286(36): 31168-79, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21757724

RESUMEN

Autoimmune thyroid diseases (AITD) arise from complex interactions between genetic, epigenetic, and environmental factors. Whole genome linkage scans and association studies have established thyroglobulin (TG) as a major AITD susceptibility gene. However, the causative TG variants and the pathogenic mechanisms are unknown. Here, we describe a genetic/epigenetic mechanism by which a newly identified TG promoter single-nucleotide polymorphism (SNP) variant predisposes to AITD. Sequencing analyses followed by case control and family-based association studies identified an SNP (-1623A→G) that was associated with AITD in the Caucasian population (p = 0.006). We show that the nucleotide substitution introduced by SNP (-1623A/G) modified a binding site for interferon regulatory factor-1 (IRF-1), a major interferon-induced transcription factor. Using chromatin immunoprecipitation, we demonstrated that IRF-1 binds to the 5' TG promoter motif, and the transcription factor binding correlates with active chromatin structure and is marked by enrichment of mono-methylated Lys-4 residue of histone H3, a signature of active transcriptional enhancers. Using reporter mutations and siRNA approaches, we demonstrate that the disease-associated allele (G) conferred increased TG promoter activity through IRF-1 binding. Finally, treatment of thyroid cells with interferon α, a known trigger of AITD, increased TG promoter activity only when it interacted with the disease-associated variant through IRF-1 binding. These results reveal a new mechanism of interaction between environmental (IFNα) and genetic (TG) factors to trigger AITD.


Asunto(s)
Autoinmunidad/genética , Epigénesis Genética , Interferón-alfa/genética , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Tiroglobulina/genética , Enfermedades de la Tiroides/genética , Sitios de Unión , Estudios de Casos y Controles , Línea Celular , Humanos , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Enfermedades de la Tiroides/inmunología
19.
Front Immunol ; 13: 1041451, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36479127

RESUMEN

CRISPR screening is a powerful tool that links specific genetic alterations to corresponding phenotypes, thus allowing for high-throughput identification of novel gene functions. Pooled CRISPR screens have enabled discovery of innate and adaptive immune response regulators in the setting of viral infection and cancer. Emerging methods couple pooled CRISPR screens with parallel high-content readouts at the transcriptomic, epigenetic, proteomic, and optical levels. These approaches are illuminating cancer immune evasion mechanisms as well as nominating novel targets that augment T cell activation, increase T cell infiltration into tumors, and promote enhanced T cell cytotoxicity. This review details recent methodological advances in high-content CRISPR screens and highlights the impact this technology is having on tumor immunology.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Proteómica , Epigenómica
20.
JCI Insight ; 7(20)2022 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-36099022

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) plays a central role in normal and aberrant wound healing, but the precise mechanism in the local environment remains elusive. Here, using a mouse model of aberrant wound healing resulting in heterotopic ossification (HO) after traumatic injury, we find autocrine TGF-ß1 signaling in macrophages, and not mesenchymal stem/progenitor cells, is critical in HO formation. In-depth single-cell transcriptomic and epigenomic analyses in combination with immunostaining of cells from the injury site demonstrated increased TGF-ß1 signaling in early infiltrating macrophages, with open chromatin regions in TGF-ß1-stimulated genes at binding sites specific for transcription factors of activated TGF-ß1 (SMAD2/3). Genetic deletion of TGF-ß1 receptor type 1 (Tgfbr1; Alk5), in macrophages, resulted in increased HO, with a trend toward decreased tendinous HO. To bypass the effect seen by altering the receptor, we administered a systemic treatment with TGF-ß1/3 ligand trap TGF-ßRII-Fc, which resulted in decreased HO formation and a delay in macrophage infiltration to the injury site. Overall, our data support the role of the TGF-ß1/ALK5 signaling pathway in HO.


Asunto(s)
Osificación Heterotópica , Factor de Crecimiento Transformador beta1 , Humanos , Cromatina/metabolismo , Ligandos , Macrófagos/metabolismo , Osificación Heterotópica/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1/metabolismo , Cicatrización de Heridas , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA