Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Immunity ; 42(4): 719-30, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25902483

RESUMEN

Within secondary lymphoid tissues, stromal reticular cells support lymphocyte function, and targeting reticular cells is a potential strategy for controlling pathogenic lymphocytes in disease. However, the mechanisms that regulate reticular cell function are not well understood. Here we found that during an immune response in lymph nodes, dendritic cells (DCs) maintain reticular cell survival in multiple compartments. DC-derived lymphotoxin beta receptor (LTßR) ligands were critical mediators, and LTßR signaling on reticular cells mediated cell survival by modulating podoplanin (PDPN). PDPN modulated integrin-mediated cell adhesion, which maintained cell survival. This DC-stromal axis maintained lymphocyte survival and the ongoing immune response. Our findings provide insight into the functions of DCs, LTßR, and PDPN and delineate a DC-stromal axis that can potentially be targeted in autoimmune or lymphoproliferative diseases.


Asunto(s)
Células Dendríticas/citología , Ganglios Linfáticos/citología , Receptor beta de Linfotoxina/inmunología , Glicoproteínas de Membrana/inmunología , Células del Estroma/citología , Animales , Adhesión Celular , Supervivencia Celular/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica , Inmunofenotipificación , Ganglios Linfáticos/inmunología , Depleción Linfocítica , Receptor beta de Linfotoxina/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal , Células del Estroma/inmunología
2.
J Immunol ; 206(2): 302-309, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33397744

RESUMEN

Photosensitivity is a sensitivity to UV radiation (UVR) commonly found in systemic lupus erythematosus (SLE) patients who have cutaneous disease. Upon even ambient UVR exposure, patients can develop inflammatory skin lesions that can reduce the quality of life. Additionally, UVR-exposed skin lesions can be associated with systemic disease flares marked by rising autoantibody titers and worsening kidney disease. Why SLE patients are photosensitive and how skin sensitivity leads to systemic disease flares are not well understood, and treatment options are limited. In recent years, the importance of immune cell-stromal interactions in tissue function and maintenance is being increasingly recognized. In this review, we discuss SLE as an anatomic circuit and review recent findings in the pathogenesis of photosensitivity with a focus on immune cell-stromal circuitry in tissue health and disease.


Asunto(s)
Lupus Eritematoso Sistémico/inmunología , Trastornos por Fotosensibilidad/inmunología , Piel/patología , Animales , Autoanticuerpos/metabolismo , Comunicación Celular , Humanos , Inmunidad Celular
3.
J Biol Chem ; 296: 100733, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33957124

RESUMEN

A disintegrin and metalloprotease 17 (ADAM17) is a cell-surface metalloprotease that serves as the principle sheddase for tumor necrosis factor α (TNFα), interleukin-6 receptor (IL-6R), and several ligands of the epidermal growth factor receptor (EGFR), regulating these crucial signaling pathways. ADAM17 activation requires its transmembrane domain, but not its cytoplasmic domain, and little is known about the role of this domain in vivo. To investigate, we used CRISPR-Cas9 to mutate the endogenous Adam17 locus in mice to produce a mutant ADAM17 lacking its cytoplasmic domain (Adam17Δcyto). Homozygous Adam17Δcyto animals were born at a Mendelian ratio and survived into adulthood with slightly wavy hair and curled whiskers, consistent with defects in ADAM17/EGFR signaling. At birth, Adam17Δcyto mice resembled Adam17-/- mice in that they had open eyes and enlarged semilunar heart valves, but they did not have bone growth plate defects. The deletion of the cytoplasmic domain resulted in strongly decreased ADAM17 protein levels in all tissues and cells examined, providing a likely cause for the hypomorphic phenotype. In functional assays, Adam17Δcyto mouse embryonic fibroblasts and bone-marrow-derived macrophages had strongly reduced ADAM17 activity, consistent with the reduced protein levels. Nevertheless, ADAM17Δcyto could be stimulated by PMA, a well-characterized posttranslational activator of ADAM17, corroborating that the cytoplasmic domain of endogenous ADAM17 is not required for its rapid response to PMA. Taken together, these results provide the first evidence that the cytoplasmic domain of ADAM17 plays a pivotal role in vivo in regulating ADAM17 levels and function.


Asunto(s)
Proteína ADAM17/química , Proteína ADAM17/metabolismo , Citoplasma/metabolismo , Proteína ADAM17/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sistemas CRISPR-Cas , Femenino , Fibroblastos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Dominios Proteicos , Estabilidad Proteica , Eliminación de Secuencia
4.
Curr Opin Rheumatol ; 34(2): 133-138, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34954700

RESUMEN

PURPOSE OF REVIEW: The aim of this review is to give insights into how novel lymphatics functions may influence autoimmunity. RECENT FINDINGS: The lymphatic system connects peripheral tissues to draining lymph nodes to regulate adaptive immunity and directly interfaces with leukocytes in lymph vessels and in the lymph node. Here, we discuss recent findings showing evidence of dysfunctional lymphatics in autoimmune disease, new understanding of how afferent lymphatic regulation can modulate immunity, lymph node lymphatic heterogeneity and how these lymphatics can directly modulate lymphocyte function, how this understanding can be harnessed for new therapeutics, and new tools for the investigation of lymphatic and immune biology. SUMMARY: Lymphatics have an active role in the regulation of inflammation and the adaptive immune response. Here, we review recent findings in lymphatics biology in peripheral tissues and lymph nodes and emphasize the relevance for better understanding autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Vasos Linfáticos , Autoinmunidad , Humanos , Ganglios Linfáticos , Sistema Linfático
5.
Curr Opin Rheumatol ; 31(6): 569-575, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31436583

RESUMEN

PURPOSE OF REVIEW: The role of type I IFNs (IFN-I) in the promotion of autoimmunity has been well established. However, its role in the skin fibrosis of systemic sclerosis (SSc) is less clear. IFN-I can participate to tissue repair, and, here, we will consider the extent to which IFN-I's role in SSc skin fibrosis may reflect in part IFN-I functions during wound healing. RECENT FINDINGS: Studies are beginning to delineate whether IFN-I has a protective or pathogenic role and how IFN-I affects tissue biology. Recent support for a pathogenic role came from a study depleting plasmacytoid dendritic cells during bleomycin-induced skin fibrosis. The depletion reduced the bleomycin-induced IFN-I-stimulated transcripts and both prevented and reversed fibrosis. Additionally, two recent articles, one identifying SSc endothelial cell injury markers and one showing repressed IFN signaling in SSc keratinocytes, suggest the possibility of unbalanced IFN-I activities on distinct cells types. SUMMARY: Recent results support a pathogenic role for IFN-I in skin fibrosis, and recent studies along with others suggest a scenario whereby SSc skin damage results from too much IFN-I-activity driving vasculopathy in combination with too little IFN-I-mediated epidermal integrity and antifibrotic fibroblast phenotype.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata , Interferón Tipo I/metabolismo , Esclerodermia Sistémica/inmunología , Piel/metabolismo , Animales , Fibroblastos/inmunología , Fibroblastos/patología , Humanos , Esclerodermia Sistémica/metabolismo , Transducción de Señal , Piel/patología
6.
Trends Immunol ; 37(11): 764-777, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27638128

RESUMEN

During normal and pathologic immune responses, lymph nodes can swell considerably. The lymph node vascular-stromal compartment supports and regulates the developing immune responses and undergoes dynamic expansion and remodeling. Recent studies have shown that dendritic cells (DCs), best known for their antigen presentation roles, can directly regulate the vascular-stromal compartment, pointing to a new perspective on DCs as facilitators of lymphoid tissue function. Here, we review the phases of lymph node vascular-stromal growth and remodeling during immune responses, discuss the roles of DCs, and discuss how this understanding can potentially be used for developing novel therapeutic approaches.


Asunto(s)
Presentación de Antígeno , Células Dendríticas/inmunología , Inmunidad , Inmunoterapia/métodos , Ganglios Linfáticos/inmunología , Células del Estroma/fisiología , Remodelación Vascular/inmunología , Animales , Antígenos/inmunología , Antígenos/metabolismo , Comunicación Celular/inmunología , Humanos , Inmunoterapia/tendencias , Péptidos/inmunología , Péptidos/metabolismo
7.
Clin Orthop Relat Res ; 477(11): 2588-2598, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31283731

RESUMEN

BACKGROUND: Periprosthetic joint infection (PJI) is one of the most devastating complications of total joint arthroplasty. Given the mortality and morbidity associated with PJI and the challenges in treating it, there has been increased interest in risk factors that can be modified before surgery. In this study, we used a novel mouse model to consider the role of the gut microbiome as a risk factor for PJI. QUESTIONS/PURPOSES: (1) Does the state of the gut microbiota before surgery influence the likelihood of developing an established infection in a mouse model of PJI? (2) How does the state of the gut microbiota before surgery influence the local and systemic response to the presence of an established infection in a mouse model of PJI? METHODS: Male C57Bl/6 mice were divided into two groups: those with modified microbiome [INCREMENT]microbiome (n = 40) and untreated mice (n = 42). In [INCREMENT]microbiome mice, the gut flora were modified using oral neomycin and ampicillin from 4 weeks to 16 weeks of age. Mice received a titanium tibial implant to mimic a joint implant and a local inoculation of Staphylococcus aureus in the synovial space (10 colony forming units [CFUs]). The proportion of animals developing an established infection in each group was determined by CFU count. The local and systemic response to established infection was determined using CFU counts in surrounding joint tissues, analysis of gait, radiographs, body weight, serum markers of inflammation, and immune cell profiles and was compared with animals that received the inoculation but resisted infection. RESULTS: A greater proportion of animals with disrupted gut microbiota had infection (29 of 40 [73%]) than did untreated animals (21 of 42 [50%]; odds ratio, 2.63, 95% CI, 1.04-6.61; p = 0.035). The immune response to established infection in mice with altered microbiota was muted; serum amyloid A, a marker of systemic infection in mice, was greater than in mice with disrupted gut microbiota with infection (689 µg/dL; range, 68-2437 µg/dL, p < 0.05); infection associated increases in monocytes and neutrophils in the spleen and local lymph node in untreated mice but not were not observed in mice with disrupted gut microbiota. CONCLUSIONS: The findings from this in vivo mouse model suggest that the gut microbiota may influence susceptibility to PJI. CLINICAL RELEVANCE: These preclinical findings support the idea that the state of the gut microbiome before surgery may influence the development of PJI and justify further preclinical and clinical studies to develop appropriate microbiome-based interventions.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Prótesis Articulares/efectos adversos , Infecciones Relacionadas con Prótesis/etiología , Infecciones Estafilocócicas/etiología , Staphylococcus aureus , Tibia/cirugía , Animales , Modelos Animales de Enfermedad , Ratones
8.
Curr Opin Rheumatol ; 29(6): 585-590, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28800024

RESUMEN

PURPOSE OF REVIEW: Dermal white adipose tissue (DWAT) is distinct from subcutaneous white adipose tissue and is lost in scleroderma skin fibrosis. The roles of DWAT loss in scleroderma skin fibrosis have not been well understood, and here we discuss recent findings that begin to provide insight into the multiple mechanisms involved. RECENT FINDINGS: The DWAT loss in part reflects the direct contribution of DWAT cells to the fibrotic tissue, with the reprogramming of adipocytes to myofibroblasts. The DWAT contains reparative adipose-derived stromal cells and expresses antifibrotic cytokines such as adiponectin, and the loss of these skin-protective mechanisms with DWAT loss further contributes to skin fibrosis and injury. SUMMARY: Potentially, halting or reversing the transdifferentiation of adipocytes to myofibroblasts along with improving survival of reparative adipose-derived stromal cells (ADSCs) and expression of antifibrotic cytokines may be effective therapeutic avenues.


Asunto(s)
Tejido Adiposo Blanco/patología , Transdiferenciación Celular/fisiología , Esclerodermia Localizada/patología , Piel/patología , Adipocitos/metabolismo , Adipocitos/patología , Adiponectina/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Citocinas/metabolismo , Dermis/metabolismo , Dermis/patología , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Esclerodermia Localizada/metabolismo , Piel/metabolismo
9.
J Immunol ; 192(9): 4153-63, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24659690

RESUMEN

Lymphadenopathy in autoimmune and other lymphoproliferative diseases is in part characterized by immunoblasts and vascular proliferation. The lymph node vasculature, along with the nonvascular stromal compartment, supports lymphocyte function, and targeting vascular-stromal expansion in inflamed nodes may modulate lymphocyte function in disease. CD11c(+) cells are essential for vascular-stromal proliferation and the upregulation of vascular endothelial growth factor (VEGF) needed for vascular proliferation. However, targetable CD11c(+) cell-derived molecular mediators, the identity of relevant CD11c(+) cells, and whether CD11c(+) cells directly stimulate VEGF-expressing stromal cells are poorly understood. In this study we show that CD11c(+) CD11b(+) CCR2-dependent monocytes and CCR7-dependent dendritic cells express IL-1ß. IL-1ß blockade, IL-1ß deficiency in radiosensitive cells, and CCR2/CCR7 double deficiency but not single deficiency all attenuate immunization-induced vascular-stromal proliferation. gp38(+) stromal fibroblastic reticular cells (FRCs) that express VEGF are enriched for Thy1(+) cells and partially overlap with CCL21-expressing FRCs, and FRC VEGF is attenuated with IL-1ß deficiency or blockade. IL-1ß localizes to the outer borders of the T zone, where VEGF-expressing cells are also enriched. Ex vivo, CD11b(+) cells enriched for IL-1ß(+) cells can directly induce cultured gp38(+)Thy1(+) FRCs to upregulate VEGF. Taken together, these results suggest a mechanism whereby multiple recruited CD11c(+) populations express IL-1ß and directly modulate FRC function to help promote the initiation of vascular-stromal growth in stimulated lymph nodes. These data provide new insight into how CD11c(+) cells regulate the lymph node vascular-stromal compartment, add to the evolving understanding of functional stromal subsets, and suggest a possible utility for IL-1ß blockade in preventing inflammatory lymph node growth.


Asunto(s)
Antígeno CD11c/metabolismo , Interleucina-1beta/metabolismo , Ganglios Linfáticos/metabolismo , Células del Estroma/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Antígeno CD11c/inmunología , Separación Celular , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interleucina-1beta/inmunología , Ganglios Linfáticos/irrigación sanguínea , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Células del Estroma/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología
10.
Curr Opin Rheumatol ; 27(6): 530-6, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26352734

RESUMEN

PURPOSE OF REVIEW: In this review of the literature from 2014 through mid-2015, we examine new data that shed light on how macrophages and other innate immune cells and signals contribute to inflammation, vascular dysfunction, and fibrosis in scleroderma. RECENT FINDINGS: Recent human studies have focused on changes early in scleroderma, and linked macrophages to inflammation in skin and progression of lung disease. Plasmacytoid dendritic cells have been implicated in vascular dysfunction. In mice, several factors have been identified that influence macrophage activation and experimental fibrosis. However, emerging data also suggest that myeloid cells can have differential effects in fibrosis. Sustained signaling through different toll-like receptors can lead to inflammation or fibrosis, and these signals can influence both immune and nonimmune cells. SUMMARY: There are many types of innate immune cells that can potentially contribute to scleroderma and will be worth exploring in detail. Experimentally dissecting the roles of macrophages based on ontogeny and activation state, and the innate signaling pathways in the tissue microenvironment, may also lead to better understanding of scleroderma pathogenesis.


Asunto(s)
Inmunidad Innata , Macrófagos/inmunología , Esclerodermia Sistémica/inmunología , Animales , Células Dendríticas/inmunología , Fibrosis/inmunología , Humanos , Inflamación/inmunología , Ratones
11.
Cell Rep ; 43(6): 114311, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38848214

RESUMEN

The lymphatic fluid is the conduit by which part of the tissue "omics" is transported to the draining lymph node for immunosurveillance. Following cannulation of the pre-nodal cervical and mesenteric afferent lymphatics, herein we investigate the lymph proteomic composition, uncovering that its composition varies according to the tissue of origin. Tissue specificity is also reflected in the dendritic cell-major histocompatibility complex class II-eluted immunopeptidome harvested from the cervical and mesenteric nodes. Following inflammatory disruption of the gut barrier, the lymph antigenic and inflammatory loads are analyzed in both mice and subjects with inflammatory bowel diseases. Gastrointestinal tissue damage reflects the lymph inflammatory and damage-associated molecular pattern signatures, microbiome-derived by-products, and immunomodulatory molecules, including metabolites of the gut-brain axis, mapped in the afferent mesenteric lymph. Our data point to the relevance of the lymphatic fluid to probe the tissue-specific antigenic and inflammatory load transported to the draining lymph node for immunosurveillance.


Asunto(s)
Antígenos , Inflamación , Ganglios Linfáticos , Linfa , Ratones Endogámicos C57BL , Animales , Ratones , Linfa/metabolismo , Linfa/inmunología , Inflamación/inmunología , Inflamación/patología , Inflamación/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Humanos , Antígenos/metabolismo , Antígenos/inmunología , Masculino , Femenino , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Inflamatorias del Intestino/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo
12.
Elife ; 132024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38860651

RESUMEN

The autoimmune disease lupus erythematosus (lupus) is characterized by photosensitivity, where even ambient ultraviolet radiation (UVR) exposure can lead to development of inflammatory skin lesions. We have previously shown that Langerhans cells (LCs) limit keratinocyte apoptosis and photosensitivity via a disintegrin and metalloprotease 17 (ADAM17)-mediated release of epidermal growth factor receptor (EGFR) ligands and that LC ADAM17 sheddase activity is reduced in lupus. Here, we sought to understand how the lupus skin environment contributes to LC ADAM17 dysfunction and, in the process, differentiate between effects on LC ADAM17 sheddase function, LC ADAM17 expression, and LC numbers. We show through transcriptomic analysis a shared IFN-rich environment in non-lesional skin across human lupus and three murine models: MRL/lpr, B6.Sle1yaa, and imiquimod (IMQ) mice. IFN-I inhibits LC ADAM17 sheddase activity in murine and human LCs, and IFNAR blockade in lupus model mice restores LC ADAM17 sheddase activity, all without consistent effects on LC ADAM17 protein expression or LC numbers. Anti-IFNAR-mediated LC ADAM17 sheddase function restoration is associated with reduced photosensitive responses that are dependent on EGFR signaling and LC ADAM17. Reactive oxygen species (ROS) is a known mediator of ADAM17 activity; we show that UVR-induced LC ROS production is reduced in lupus model mice, restored by anti-IFNAR, and is cytoplasmic in origin. Our findings suggest that IFN-I promotes photosensitivity at least in part by inhibiting UVR-induced LC ADAM17 sheddase function and raise the possibility that anifrolumab ameliorates lupus skin disease in part by restoring this function. This work provides insight into IFN-I-mediated disease mechanisms, LC regulation, and a potential mechanism of action for anifrolumab in lupus.


Asunto(s)
Proteína ADAM17 , Células de Langerhans , Lupus Eritematoso Sistémico , Piel , Proteína ADAM17/metabolismo , Proteína ADAM17/genética , Animales , Humanos , Células de Langerhans/metabolismo , Ratones , Piel/metabolismo , Piel/patología , Piel/efectos de la radiación , Lupus Eritematoso Sistémico/metabolismo , Rayos Ultravioleta/efectos adversos , Femenino , Modelos Animales de Enfermedad , Trastornos por Fotosensibilidad/metabolismo , Interferones/metabolismo , Ratones Endogámicos MRL lpr
13.
J Immunol ; 187(11): 5558-67, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22031764

RESUMEN

Lymph node blood vessels play important roles in the support and trafficking of immune cells. The blood vasculature is a component of the vascular-stromal compartment that also includes the lymphatic vasculature and fibroblastic reticular cells (FRCs). During immune responses as lymph nodes swell, the blood vasculature undergoes a rapid proliferative growth that is initially dependent on CD11c(+) cells and vascular endothelial growth factor (VEGF) but is independent of lymphocytes. The lymphatic vasculature grows with similar kinetics and VEGF dependence, suggesting coregulation of blood and lymphatic vascular growth, but lymphatic growth has been shown to be B cell dependent. In this article, we show that blood vascular, lymphatic, and FRC growth are coordinately regulated and identify two distinct phases of vascular-stromal growth--an initiation phase, characterized by upregulated vascular-stromal proliferation, and a subsequent expansion phase. The initiation phase is CD11c(+) cell dependent and T/B cell independent, whereas the expansion phase is dependent on B and T cells together. Using CCR7(-/-) mice and selective depletion of migratory skin dendritic cells, we show that endogenous skin-derived dendritic cells are not important during the initiation phase and uncover a modest regulatory role for CCR7. Finally, we show that FRC VEGF expression is upregulated during initiation and that dendritic cells can stimulate increased fibroblastic VEGF, suggesting the scenario that lymph node-resident CD11c(+) cells orchestrate the initiation of blood and lymphatic vascular growth in part by stimulating FRCs to upregulate VEGF. These results illustrate how the lymph node microenvironment is shaped by the cells it supports.


Asunto(s)
Linfocitos B/inmunología , Microambiente Celular/inmunología , Quimiotaxis de Leucocito/inmunología , Células Dendríticas/inmunología , Ganglios Linfáticos/irrigación sanguínea , Linfocitos T/inmunología , Animales , Antígeno CD11c/inmunología , Separación Celular , Células Endoteliales/inmunología , Fibroblastos/inmunología , Citometría de Flujo , Ganglios Linfáticos/inmunología , Vasos Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Factor A de Crecimiento Endotelial Vascular/inmunología
14.
Cardiovasc Res ; 119(2): 506-519, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35815623

RESUMEN

AIMS: Growing evidence correlate the accrual of the sphingolipid ceramide in plasma and cardiac tissue with heart failure (HF). Regulation of sphingolipid metabolism in the heart and the pathological impact of its derangement remain poorly understood. Recently, we discovered that Nogo-B, a membrane protein of endoplasmic reticulum, abundant in the vascular wall, down-regulates the sphingolipid de novo biosynthesis via serine palmitoyltransferase (SPT), first and rate liming enzyme, to impact vascular functions and blood pressure. Nogo-A, a splice isoform of Nogo, is transiently expressed in cardiomyocyte (CM) following pressure overload. Cardiac Nogo is up-regulated in dilated and ischaemic cardiomyopathies in animals and humans. However, its biological function in the heart remains unknown. METHODS AND RESULTS: We discovered that Nogo-A is a negative regulator of SPT activity and refrains ceramide de novo biosynthesis in CM exposed to haemodynamic stress, hence limiting ceramide accrual. At 7 days following transverse aortic constriction (TAC), SPT activity was significantly up-regulated in CM lacking Nogo-A and correlated with ceramide accrual, particularly very long-chain ceramides, which are the most abundant in CM, resulting in the suppression of 'beneficial' autophagy. At 3 months post-TAC, mice lacking Nogo-A in CM showed worse pathological cardiac hypertrophy and dysfunction, with ca. 50% mortality rate. CONCLUSION: Mechanistically, Nogo-A refrains ceramides from accrual, therefore preserves the 'beneficial' autophagy, mitochondrial function, and metabolic gene expression, limiting the progression to HF under sustained stress.


Asunto(s)
Insuficiencia Cardíaca , Esfingolípidos , Humanos , Ratones , Animales , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Esfingolípidos/metabolismo , Ceramidas/metabolismo , Insuficiencia Cardíaca/genética , Miocitos Cardíacos/metabolismo
15.
J Exp Med ; 203(8): 1903-13, 2006 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-16831898

RESUMEN

Lymph nodes grow rapidly and robustly at the initiation of an immune response, and this growth is accompanied by growth of the blood vessels. Although the vessels are critical for supplying nutrients and for controlling cell trafficking, the regulation of lymph node vascular growth is not well understood. We show that lymph node endothelial cells begin to proliferate within 2 d of immunization and undergo a corresponding expansion in cell numbers. Endothelial cell proliferation is dependent on CD11c+ dendritic cells (DCs), and the subcutaneous injection of DCs is sufficient to trigger endothelial cell proliferation and growth. Lymph node endothelial cell proliferation is dependent on vascular endothelial growth factor (VEGF), and DCs are associated with increased lymph node VEGF levels. DC-induced endothelial cell proliferation and increased VEGF levels are mediated by DC-induced recruitment of blood-borne cells. Vascular growth in the draining lymph node includes the growth of high endothelial venule endothelial cells and is functionally associated with increased cell entry into the lymph node. Collectively, our results suggest a scenario whereby endothelial cell expansion in the draining lymph node is induced by DCs as part of a program that optimizes the microenvironment for the ensuing immune response.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Células Dendríticas/fisiología , Ganglios Linfáticos/irrigación sanguínea , Animales , Antígeno CD11c/inmunología , Proliferación Celular , Células Endoteliales/citología , Proteínas de Homeodominio/metabolismo , Inmunización , Ganglios Linfáticos/citología , Vasos Linfáticos/citología , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Clin Immunol ; 144(2): 109-16, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22717771

RESUMEN

Lymphadenopathy occurs in many autoimmune and inflammatory diseases, and vascular proliferation is a common feature in the enlarged lymph nodes. The lymph node vasculature plays critical roles in delivering immune cells as well as oxygen and micronutrients, and therefore represents a potential target for therapeutic manipulation of immunity. In this review, we discuss recent insights made in understanding the growth and function of the vascular and associated stromal compartment in immune-stimulated lymph nodes and the potential utility of altering this process in autoimmune diseases.


Asunto(s)
Ganglios Linfáticos/irrigación sanguínea , Ganglios Linfáticos/inmunología , Vasos Linfáticos/citología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Vasos Sanguíneos/citología , Células Endoteliales/metabolismo , Humanos , Células del Estroma/citología
17.
Curr Rheumatol Rep ; 14(1): 30-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22006170

RESUMEN

Dendritic cells are professional antigen-presenting cells that are most studied for their function in mediating T-cell tolerance and T-cell activation. In addition, recent evidence indicates that dendritic cells can regulate the vasculature and function of fibroblast-type cells. The potential contribution of dendritic cells to scleroderma and fibrosis is not well-understood. In this article, we review recent studies as well as describe our own ongoing work that points toward a role for dendritic cells in scleroderma and fibrosis.


Asunto(s)
Células Dendríticas/fisiología , Riñón/patología , Hígado/patología , Pulmón/patología , Esclerodermia Sistémica/fisiopatología , Piel/patología , Fibrosis , Humanos
18.
J Immunol ; 184(8): 4247-57, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20231692

RESUMEN

Lymph node expansion during immune responses is accompanied by rapid vascular expansion. The re-establishment of quiescence and stabilization of the newly expanded vasculature and the regulatory mechanisms involved have not been well studied. We show that although initiation of vascular expansion in immune-stimulated nodes is associated with upregulated endothelial cell proliferation, increased high endothelial venule trafficking efficiency and VCAM-1 expression, and disrupted perivascular fibroblastic reticular cell organization, the re-establishment of vascular quiescence and stabilization postexpansion is characterized by reversal of these phenomena. Although CD11c(med) cells are associated with the initiation of vascular expansion, CD11c(hi)MHC class II (MHC II)(med) dendritic cells (DCs) accumulate later, and their short-term depletion in mice abrogates the re-establishment of vascular quiescence and stabilization. CD11c(hi)MHC II(med) cells promote endothelial cell quiescence in vitro and, in vivo, mediate quiescence at least in part by mediating reduced lymph node vascular endothelial growth factor. Disrupted vascular quiescence and stabilization in expanded nodes is associated with attenuated T cell-dependent B cell responses. These results describe a novel mechanism whereby CD11c(hi)MHC II(med) DCs regulate the re-establishment of vascular quiescence and stabilization after lymph node vascular expansion and suggest that these DCs function in part to orchestrate the microenvironmental alterations required for successful immunity.


Asunto(s)
Antígeno CD11c/fisiología , Células Dendríticas/inmunología , Endotelio Vascular/inmunología , Antígenos de Histocompatibilidad Clase II/biosíntesis , Ganglios Linfáticos/irrigación sanguínea , Ganglios Linfáticos/inmunología , Vasos Linfáticos/inmunología , Activación de Linfocitos/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígeno CD11c/biosíntesis , Línea Celular Tumoral , Movimiento Celular/inmunología , Proliferación Celular , Células Cultivadas , Células Dendríticas/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Fibroblastos/inmunología , Fibroblastos/metabolismo , Ganglios Linfáticos/citología , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transporte de Proteínas/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Regulación hacia Arriba/inmunología
19.
J Rheumatol ; 49(9): 1026-1030, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35649551

RESUMEN

OBJECTIVE: In antiphospholipid antibody (aPL) nephropathy, activation of the mammalian target of rapamycin (mTOR) contributes to endothelial cell proliferation, a key finding of aPL microvascular disease. Here, we examined mTOR activation in the skin of aPL-positive patients with livedo. METHODS: Three patient groups with livedo were studied: (1) persistently aPL-positive with systemic lupus erythematosus (SLE); (2) persistently aPL-positive without SLE; and (3) aPL-negative SLE (control). After collecting aPL-related medical history, two 5-mm skin biopsies of livedo were performed on each patient: (1) peripheral (erythematous-violaceous lesion); and (2) central (nonviolaceous area). We stained specimens for phosphorylated protein kinase B (p-AKT) and phosphorylated S6 ribosomal protein (p-S6RP) as mTOR activity markers, CD31 to identify endothelial cells, and Ki-67 to show cellular proliferation. We counted cells in the epidermis and compared mTOR-positive cell counts between peripheral and central samples, and between patient groups, using Freidman test and Wilcoxon signed-rank test. RESULTS: Ten patients with livedo reticularis were enrolled: 4 aPL-positive without SLE (antiphospholipid syndrome [APS] classification met, n = 3), 4 aPL-positive SLE (APS classification met, n = 3), and 2 aPL-negative SLE (control). In all aPL-positive patients, epidermal p-AKT and p-S6RP staining were significantly increased in both peripheral and central skin samples when compared to aPL-negative SLE controls; both were more pronounced in the lower basal layers of epidermis. CONCLUSION: Our study demonstrates increased mTOR activity in livedoid lesions of aPL-positive patients with or without SLE compared to aPL-negative patients with SLE, with more prominent activity in the lower basal layers of the epidermis. These findings may serve as a basis for further investigating the mTOR pathway in aPL-positive patients.


Asunto(s)
Síndrome Antifosfolípido , Livedo Reticularis , Lupus Eritematoso Sistémico , Serina-Treonina Quinasas TOR , Humanos , Anticuerpos Antifosfolípidos , Células Endoteliales , Antígeno Ki-67 , Proteínas Proto-Oncogénicas c-akt , Proteínas Ribosómicas , Sirolimus
20.
J Exp Med ; 218(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34374712

RESUMEN

In this issue of JEM, Podstawka et al. (2021. J. Exp. Med.https://doi.org/10.1084/jem.20210409) show that B cells can limit neutrophil responses within the lung microvasculature by marginating and acting on marginated neutrophils. This study provides a new view of B cells and reveals a novel mechanism of cell-mediated intravascular regulation.


Asunto(s)
Linfocitos B , Neutrófilos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA