Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Cell ; 185(10): 1694-1708.e19, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35447074

RESUMEN

Immunotherapy is a promising treatment for triple-negative breast cancer (TNBC), but patients relapse, highlighting the need to understand the mechanisms of resistance. We discovered that in primary breast cancer, tumor cells that resist T cell attack are quiescent. Quiescent cancer cells (QCCs) form clusters with reduced immune infiltration. They also display superior tumorigenic capacity and higher expression of chemotherapy resistance and stemness genes. We adapted single-cell RNA-sequencing with precise spatial resolution to profile infiltrating cells inside and outside the QCC niche. This transcriptomic analysis revealed hypoxia-induced programs and identified more exhausted T cells, tumor-protective fibroblasts, and dysfunctional dendritic cells inside clusters of QCCs. This uncovered differential phenotypes in infiltrating cells based on their intra-tumor location. Thus, QCCs constitute immunotherapy-resistant reservoirs by orchestrating a local hypoxic immune-suppressive milieu that blocks T cell function. Eliminating QCCs holds the promise to counteract immunotherapy resistance and prevent disease recurrence in TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Inmunosupresores/uso terapéutico , Inmunoterapia , Recurrencia Local de Neoplasia , Linfocitos T/patología , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral
2.
Trends Immunol ; 36(10): 578-604, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26431937

RESUMEN

The nervous system and the immune system are the principal sensory interfaces between the internal and external environment. They are responsible for recognizing, integrating, and responding to varied stimuli, and have the capacity to form memories of these encounters leading to learned or 'adaptive' future responses. We review current understanding of the cross-regulation between these systems. The autonomic and somatosensory nervous systems regulate both the development and deployment of immune cells, with broad functions that impact on hematopoiesis as well as on priming, migration, and cytokine production. In turn, specific immune cell subsets contribute to homeostatic neural circuits such as those controlling metabolism, hypertension, and the inflammatory reflex. We examine the contribution of the somatosensory system to autoimmune, autoinflammatory, allergic, and infectious processes in barrier tissues and, in this context, discuss opportunities for therapeutic manipulation of neuro-immune interactions.


Asunto(s)
Homeostasis , Neuroinmunomodulación , Neuronas/metabolismo , Sistema Nervioso Periférico/inmunología , Sistema Nervioso Periférico/metabolismo , Animales , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/fisiología , Sistema Nervioso Periférico/fisiología , Transducción de Señal
3.
BMC Biol ; 15(1): 45, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28526034

RESUMEN

BACKGROUND: Intravascular leukocyte recruitment in most vertebrate tissues is restricted to postcapillary and collecting venules, whereas capillaries and arterioles usually support little or no leukocyte adhesion. This segmental restriction is thought to be mediated by endothelial, rather than hemodynamic, differences. The underlying mechanisms are largely unknown, in part because effective tools to distinguish, isolate, and analyze venular endothelial cells (V-ECs) and non-venular endothelial cells (NV-ECs) have been unavailable. We hypothesized that the atypical chemokine receptor DARC (Duffy Antigen Receptor for Chemokines, a.k.a. ACKR1 or CD234) may distinguish V-ECs versus NV-ECs in mice. METHODS: We generated a rat-anti-mouse monoclonal antibody (MAb) that specifically recognizes the erythroid and endothelial forms of native, surface-expressed DARC. Using this reagent, we characterized DARC expression and distribution in the microvasculature of murine tissues. RESULTS: DARC was exquisitely restricted to post-capillary and small collecting venules and completely absent from arteries, arterioles, capillaries, veins, and most lymphatics in every tissue analyzed. Accordingly, intravital microscopy showed that adhesive leukocyte-endothelial interactions were restricted to DARC+ venules. DARC was detectable over the entire circumference of V-ECs, but was more concentrated at cell-cell junctions. Analysis of single-cell suspensions suggested that the frequency of V-ECs among the total microvascular EC pool varies considerably between different tissues. CONCLUSIONS: Immunostaining of endothelial DARC allows the identification and isolation of intact V-ECs from multiple murine tissues. This strategy may be useful to dissect the mechanisms underlying segmental microvascular specialization in healthy and diseased tissues and to characterize the role of EC subsets in tissue-homeostasis, immune surveillance, infection, inflammation, and malignancies.


Asunto(s)
Sistema del Grupo Sanguíneo Duffy , Células Endoteliales , Endotelio Vascular , Regulación de la Expresión Génica , Ratones , Receptores de Superficie Celular , Animales , Ratones/genética , Ratones/metabolismo , Sistema del Grupo Sanguíneo Duffy/genética , Sistema del Grupo Sanguíneo Duffy/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Venas/metabolismo
4.
J Cell Sci ; 127(Pt 17): 3768-81, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24994937

RESUMEN

Cell-cell adhesions are important sites through which cells experience and resist forces. In endothelial cells, these forces regulate junction dynamics and determine endothelial barrier strength. We identify the Ig superfamily member EMMPRIN (also known as basigin) as a coordinator of forces at endothelial junctions. EMMPRIN localization at junctions correlates with endothelial junction strength in different mouse vascular beds. Accordingly, EMMPRIN-deficient mice show altered junctions and increased junction permeability. Lack of EMMPRIN alters the localization and function of VE-cadherin (also known as cadherin-5) by decreasing both actomyosin contractility and tugging forces at endothelial cell junctions. EMMPRIN ensures proper actomyosin-driven maturation of competent endothelial junctions by forming a molecular complex with γ-catenin (also known as junction plakoglobin) and Nm23 (also known as NME1), a nucleoside diphosphate kinase, thereby locally providing ATP to fuel the actomyosin machinery. These results provide a novel mechanism for the regulation of actomyosin contractility at endothelial junctions and might have broader implications in biological contexts such as angiogenesis, collective migration and tissue morphogenesis by coupling compartmentalized energy production to junction assembly.


Asunto(s)
Actomiosina/metabolismo , Adenosina Trifosfato/biosíntesis , Basigina/metabolismo , Células Endoteliales/citología , Nucleósido Difosfato Quinasas NM23/metabolismo , gamma Catenina/metabolismo , Animales , Adhesión Celular/fisiología , Membrana Celular/metabolismo , Células Cultivadas , Proteínas del Citoesqueleto/biosíntesis , Endotelio Vascular/metabolismo , Uniones Intercelulares/metabolismo , Ratones
5.
J Immunol ; 193(6): 3023-35, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25114107

RESUMEN

Adrenergic receptors are expressed on the surface of inflammation-mediating cells, but their potential role in the regulation of the inflammatory response is still poorly understood. The objectives of this work were to study the effects of α2-adrenergic agonists on the inflammatory response in vivo and to determine their mechanism of action. In two mouse models of inflammation, zymosan air pouch and thioglycolate-induced peritonitis models, the i.m. treatment with xylazine or UK14304, two α2-adrenergic agonists, reduced neutrophil migration by 60%. The α2-adrenergic antagonist RX821002 abrogated this effect. In flow cytometry experiments, the basal surface expression of L-selectin and CD11b was modified neither in murine nor in human neutrophils upon α2-agonist treatment. Similar experiments in HUVEC showed that UK14304 prevented the activation-dependent upregulation of ICAM-1. In contrast, UK14304 augmented electrical resistance and reduced macromolecular transport through a confluent HUVEC monolayer. In flow chamber experiments, under postcapillary venule-like flow conditions, the pretreatment of HUVECs, but not neutrophils, with α2-agonists decreased transendothelial migration, without affecting neutrophil rolling. Interestingly, α2-agonists prevented the TNF-α-mediated decrease in expression of the adherens junctional molecules, VE-cadherin, ß-catenin, and plakoglobin, and reduced the ICAM-1-mediated phosphorylation of VE-cadherin by immunofluorescence and confocal analysis and Western blot analysis, respectively. These findings indicate that α2-adrenoceptors trigger signals that protect the integrity of endothelial adherens junctions during the inflammatory response, thus pointing at the vascular endothelium as a therapeutic target for the management of inflammatory processes in humans.


Asunto(s)
Uniones Adherentes/inmunología , Endotelio Vascular/inmunología , Neutrófilos/inmunología , Receptores Adrenérgicos alfa 2/inmunología , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Animales , Antígenos CD/biosíntesis , Tartrato de Brimonidina , Antígeno CD11b/biosíntesis , Cadherinas/biosíntesis , Humanos , Idazoxan/análogos & derivados , Idazoxan/farmacología , Inflamación/inmunología , Molécula 1 de Adhesión Intercelular/biosíntesis , Selectina L/biosíntesis , Masculino , Ratones , Peritonitis/inducido químicamente , Quinoxalinas/farmacología , Receptores Adrenérgicos alfa 2/biosíntesis , Tioglicolatos/farmacología , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Regulación hacia Arriba/efectos de los fármacos , Xilazina/farmacología , Zimosan/farmacología , beta Catenina/biosíntesis , gamma Catenina/biosíntesis
6.
Eur J Immunol ; 43(1): 55-64, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22975861

RESUMEN

Diphenylamine-based nonsteroidal antiinflammatory drugs (NSAIDs) are able to cause in vitro the shedding of L-selectin. The aim of this work was to determine the physio-logic relevance of L-selectin shedding in the antiinflammatory effect exerted by NSAIDs in vivo. Chemical compounds structurally related to NSAIDs - including diphenyl-amine, N-phenylanthranilic acid (N-Ph), diphenylacetic acid - as well as the traditional NSAID indomethacin were studied using the zymosan air-pouch mouse model. Animals intramuscularly pretreated with indomethacin or N-Ph, but not with diphenyl-amine or diphenylacetic acid, showed a significant dose-dependent reduction in the number of neutrophils compared with untreated animals (N-Ph, IC50 = 6.7 mg/kg). Except for indomethacin, none of these compounds caused any significant reduction in cyclooxygenase-1 activity in vivo. In flow chamber experiments, N-Ph reduced the capability of human neutrophils to pass across the endothelial barrier by interfering with leukocyte rolling step on HUVEC. N-Ph, but not diphenylacetic acid, induced activation-independent L-selectin shedding in mouse neutrophils. Interestingly, N-Ph exerted an antiinflammatory effect similar to that of the anti-L-selectin blocking antibody Mel-14, although no additive action was observed when both compounds were combined. These data suggest that the L-selectin shedding induced by NSAIDs may be involved in the antiinflammatory action exerted by these compounds in clinical settings.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Difenilamina/administración & dosificación , Células Endoteliales de la Vena Umbilical Humana/inmunología , Selectina L/metabolismo , Neutrófilos/efectos de los fármacos , Animales , Línea Celular , Ciclooxigenasa 1/metabolismo , Difenilamina/análogos & derivados , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Inflamación/inmunología , Recuento de Leucocitos , Rodamiento de Leucocito/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos , Neutrófilos/inmunología
7.
J Autoimmun ; 55: 51-62, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24934597

RESUMEN

Although FoxP3(+) regulatory T cells are key players in the maintenance of immune tolerance and autoimmunity, the lack of specific markers constitute an obstacle to their use for immunotherapy protocols. In this study, we have investigated the role of the C-type lectin receptor CD69 in the suppressor function of Tregs and maintenance of immune tolerance towards harmless inhaled antigens. We identified a novel FoxP3(+)CD69(+) Treg subset capable to maintain immune tolerance and protect to developing inflammation. Although CD69(+) and CD69(-)FoxP3(+) Tregs exist in homeostasis, only CD69-expressing Tregs express high levels of CTLA-4, ICOS, CD38 and GITR suppression-associated markers, secrete high amounts of TGFß and have potent suppressor activity. This activity is regulated by STAT5 and ERK signaling pathways and is impaired by antibody-mediated down-regulation of CD69 expression. Moreover, immunotherapy with FoxP3(+)CD69(+) Tregs restores the homeostasis in Cd69(-/-) mice, that fail to induce tolerance, and is also highly proficient in the prevention of inflammation. The identification of the FoxP3(+)CD69(+) Treg subset paves the way toward the development of new therapeutic strategies to control immune homeostasis and autoimmunity.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica/fisiología , Lectinas Tipo C/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD/genética , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación de Linfocitos T/genética , Regulación de la Expresión Génica/genética , Lectinas Tipo C/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Linfocitos T Reguladores/citología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
8.
J Immunol ; 189(11): 5402-10, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23089395

RESUMEN

Given that TLRs and sphingosine-1-phosphate (S1P) are key players in inflammation, we explored the potential interplay between TLRs and S1P in the adhesion/inflammatory pathways in primary human endothelial cells. As determined by Western blot and flow cytometry, cells treated with LPS (a TLR4 ligand) and S1P showed significantly enhanced expression of adhesion molecules such as ICAM-1 and E-selectin compared with the effect of either ligand alone. Cell-type differences on E-selectin upregulation were observed. In contrast, no cooperation effect on ICAM-1 or E-selectin was observed with a TLR2/TLR1 ligand. Consistent with an increase in adhesion molecule expression, endothelial cell treatment with LPS plus S1P significantly enhanced adhesion of PBMCs under shear stress conditions compared with the effect of either ligand alone and exhibited comparable levels of cell adhesion strength as those after TNF-α treatment. Moreover, LPS and S1P cooperated to increase the expression of proinflammatory molecules such as IL-6, cyclooxygenase-2, and prostacyclin, as determined by ELISA and Western blot. The analysis of signaling pathways revealed the synergistic phosphorylation of ERK upon LPS plus S1P treatment of HUVEC and human aortic endothelial cells and cell-type differences on p38 and NF-κB activation. Moreover, pharmacological and small interfering RNA experiments disclosed the involvement of S1P(1/3) and NF-κB in the cooperation effect and that cell origin determines the S1P receptors and signaling routes involved. Sphingosine kinase activity induction upon LPS plus S1P treatment suggests S1P- Sphingosine kinase axis involvement. In summary, LPS and S1P cooperate to increase proinflammatory molecules in endothelial cells and, in turn, to augment leukocyte adhesion, thus exacerbating S1P-mediated proadhesive/proinflammatory properties.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Inflamación/inmunología , Lipopolisacáridos/farmacología , Lisofosfolípidos/farmacología , Esfingosina/análogos & derivados , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/inmunología , Sinergismo Farmacológico , Selectina E/genética , Selectina E/inmunología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Epoprostenol/genética , Epoprostenol/inmunología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/genética , Inflamación/patología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , FN-kappa B/genética , FN-kappa B/inmunología , Especificidad de Órganos , Transducción de Señal/efectos de los fármacos , Esfingosina/farmacología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
9.
Elife ; 122023 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-37534876

RESUMEN

Background: Although there are several efficacious vaccines against COVID-19, vaccination rates in many regions around the world remain insufficient to prevent continued high disease burden and emergence of viral variants. Repurposing of existing therapeutics that prevent or mitigate severe COVID-19 could help to address these challenges. The objective of this study was to determine whether prior use of bisphosphonates is associated with reduced incidence and/or severity of COVID-19. Methods: A retrospective cohort study utilizing payer-complete health insurance claims data from 8,239,790 patients with continuous medical and prescription insurance January 1, 2019 to June 30, 2020 was performed. The primary exposure of interest was use of any bisphosphonate from January 1, 2019 to February 29, 2020. Bisphosphonate users were identified as patients having at least one bisphosphonate claim during this period, who were then 1:1 propensity score-matched to bisphosphonate non-users by age, gender, insurance type, primary-care-provider visit in 2019, and comorbidity burden. Main outcomes of interest included: (a) any testing for SARS-CoV-2 infection; (b) COVID-19 diagnosis; and (c) hospitalization with a COVID-19 diagnosis between March 1, 2020 and June 30, 2020. Multiple sensitivity analyses were also performed to assess core study outcomes amongst more restrictive matches between BP users/non-users, as well as assessing the relationship between BP-use and other respiratory infections (pneumonia, acute bronchitis) both during the same study period as well as before the COVID outbreak. Results: A total of 7,906,603 patients for whom continuous medical and prescription insurance information was available were selected. A total of 450,366 bisphosphonate users were identified and 1:1 propensity score-matched to bisphosphonate non-users. Bisphosphonate users had lower odds ratios (OR) of testing for SARS-CoV-2 infection (OR = 0.22; 95%CI:0.21-0.23; p<0.001), COVID-19 diagnosis (OR = 0.23; 95%CI:0.22-0.24; p<0.001), and COVID-19-related hospitalization (OR = 0.26; 95%CI:0.24-0.29; p<0.001). Sensitivity analyses yielded results consistent with the primary analysis. Bisphosphonate-use was also associated with decreased odds of acute bronchitis (OR = 0.23; 95%CI:0.22-0.23; p<0.001) or pneumonia (OR = 0.32; 95%CI:0.31-0.34; p<0.001) in 2019, suggesting that bisphosphonates may protect against respiratory infections by a variety of pathogens, including but not limited to SARS-CoV-2. Conclusions: Prior bisphosphonate-use was associated with dramatically reduced odds of SARS-CoV-2 testing, COVID-19 diagnosis, and COVID-19-related hospitalizations. Prospective clinical trials will be required to establish a causal role for bisphosphonate-use in COVID-19-related outcomes. Funding: This study was supported by NIH grants, AR068383 and AI155865, a grant from MassCPR (to UHvA) and a CRI Irvington postdoctoral fellowship, CRI2453 (to PH).


The COVID-19 pandemic challenged the world to rapidly develop strategies to combat the virus responsible for the disease. While several effective vaccines and new drugs have since become available, these therapies are not always easy to access and take time to generate and distribute. To address these challenges, researchers have tried to find ways to repurpose existing medications that are already commonly used and known to be safe. One potential candidate are bisphosphonates, a family of drugs used to reduce bone loss in patients with osteoporosis. Bisphosphonates have been shown to boost the immune response to viral infections, and it has been observed that patients prescribed these drugs are less likely to develop or die from pneumonia. But whether bisphosphonates are effective against COVID-19 had not been fully explored. To investigate, Thompson, Wang et al. analyzed insurance claims data from about 8 million patients between January 2019 and June 2020, including around 450,000 individuals that had filled a prescription for bisphosphonates. Patients prescribed bisphosphonates were then compared to non-users that were similar in terms of their gender, age, the type of health insurance they had, their access to healthcare, and other health comorbidities. The study revealed that bisphosphonate users were around three to five times less likely to be tested for, diagnosed with, or hospitalized for COVID-19 during the first four months of the pandemic. They were also less commonly diagnosed with other respiratory infections in 2019, like bronchitis or pneumonia. Although the results suggest that bisphosphonates provide some protection against COVID-19, they cannot directly prove it. Verifying that bisphosphonates can treat or prevent COVID-19 and/or other respiratory infections requires more studies that follow patients in real-time rather than studying previously collected data. If such studies confirm the link, bisphosphonates could be a helpful tool to protect against COVID-19 or other virus outbreaks. The drugs are widely available, safe, and affordable, and therefore may provide an alternative for patients who cannot access other medications or vaccines.


Asunto(s)
Bronquitis , COVID-19 , Infecciones del Sistema Respiratorio , Humanos , COVID-19/epidemiología , Difosfonatos/uso terapéutico , Prueba de COVID-19 , SARS-CoV-2 , Estudios Retrospectivos , Vacunas contra la COVID-19 , Estudios Prospectivos , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Bronquitis/tratamiento farmacológico
10.
JCI Insight ; 8(24)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38131378

RESUMEN

Secondary lymphedema occurs in up to 20% of patients after lymphadenectomy performed for the surgical management of tumors involving the breast, prostate, uterus, and skin. Patients develop progressive edema of the affected extremity due to retention of protein-rich lymphatic fluid. Despite compression therapy, patients progress to chronic lymphedema in which noncompressible fibrosis and adipose tissue are deposited within the extremity. The presence of fibrosis led to our hypothesis that rosiglitazone, a PPARγ agonist that inhibits fibrosis, would reduce fibrosis in a mouse model of secondary lymphedema after hind limb lymphadenectomy. In vivo, rosiglitazone reduced fibrosis in the hind limb after lymphadenectomy. Our findings verified that rosiglitazone reestablished the adipogenic features of TGF-ß1-treated mesenchymal cells in vitro. Despite this, rosiglitazone led to a reduction in adipose tissue deposition. Single-cell RNA-Seq data obtained from human tissues and flow cytometric and histological evaluation of mouse tissues demonstrated increased presence of PDGFRα+ cells in lymphedema; human tissue analysis verified these cells have the capacity for adipogenic and fibrogenic differentiation. Upon treatment with rosiglitazone, we noted a reduction in the overall quantity of PDGFRα+ cells and LipidTOX+ cells. Our findings provide a framework for treating secondary lymphedema as a condition of fibrosis and adipose tissue deposition, both of which, paradoxically, can be prevented with a pro-adipogenic agent.


Asunto(s)
Linfedema , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Masculino , Femenino , Humanos , Ratones , Animales , PPAR gamma , Rosiglitazona/farmacología , Rosiglitazona/uso terapéutico , Linfedema/tratamiento farmacológico , Fibrosis
11.
EMBO J ; 27(8): 1206-18, 2008 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-18369319

RESUMEN

Cell migration requires integration of signals arising from both the extracellular matrix and messengers acting through G protein-coupled receptors (GPCRs). We find that increased levels of G protein-coupled receptor kinase 2 (GRK2), a key player in GPCR regulation, potentiate migration of epithelial cells towards fibronectin, whereas such process is decreased in embryonic fibroblasts from hemizygous GRK2 mice or upon knockdown of GRK2 expression. Interestingly, the GRK2 effect on fibronectin-mediated cell migration involves the paracrine/autocrine activation of a sphingosine-1-phosphate (S1P) Gi-coupled GPCR. GRK2 positively modulates the activity of the Rac/PAK/MEK/ERK pathway in response to adhesion and S1P by a mechanism involving the phosphorylation-dependent, dynamic interaction of GRK2 with GIT1, a key scaffolding protein in cell migration processes. Furthermore, decreased GRK2 levels in hemizygous mice result in delayed wound healing rate in vivo, consistent with a physiological role of GRK2 as a regulator of coordinated integrin and GPCR-directed epithelial cell migration.


Asunto(s)
Movimiento Celular , Células Epiteliales/citología , Células Epiteliales/fisiología , Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Animales , Células COS , Línea Celular , Línea Celular Transformada , Movimiento Celular/genética , Chlorocebus aethiops , Fibronectinas/fisiología , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Células HeLa , Humanos , Integrinas/fisiología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo
12.
STAR Protoc ; 3(4): 101795, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36325581

RESUMEN

Single-cell techniques have revolutionized biology; however, the required sample processing inherently implies the loss of spatial localization. Here, using an approach called photoconversion of areas to dissect micro-environments (PADME), we detail steps to isolate live single cells from a primary breast tumor while retaining spatial information by combining cell photolabeling and FACS (fluorescence-activated cell sorting). These live cells can be subsequently used for myriad techniques, from flow cytometry to single-cell RNA sequencing or other single cell "omics" approach. For complete details on the use and execution of this protocol, please refer to Baldominos et al. (2022).


Asunto(s)
Citometría de Flujo , Citometría de Flujo/métodos
13.
Circulation ; 122(14): 1396-404, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20855659

RESUMEN

BACKGROUND: Experimental autoimmune myocarditis (EAM), a mouse model of post-infectious cardiomyopathy, reflects mechanisms of inflammatory cardiomyopathy in humans. EAM is characterized by an infiltration of inflammatory cells into the myocardium that can be followed by myocyte fibrosis, edema, and necrosis, leading to ventricular wall dysfunction and heart failure. Different data indicate that CD69 exerts an important immunoregulatory effect in vivo. However, the possible role of CD69 in autoimmune myocarditis has not been studied. METHODS AND RESULTS: We have explored the role of the leukocyte regulatory molecule CD69 in the inflammation that leads to cardiac dysfunction after myocardial injury in EAM. We have found that after induction of EAM, the draining lymph nodes from CD69-deficient mice developed an exacerbated Th17 inflammatory response, resulting in increases in the numbers of infiltrating leukocytes in the myocardium. In the chronic phase of EAM, transthoracic echocardiography revealed a significantly reduced left ventricular fractional shortening and a decreased ejection fraction in CD69-deficient mice, indicative of an impaired cardiac contractility. This condition was accompanied by a greater extent of myocardial fibrosis, an elevated number of sinus pauses on ECG, and an enhanced ratio of heart weight to body weight in CD69-/- mice. Moreover, both bone marrow transplantation and adoptive transfer of Th17 cells isolated from immunized CD69-/- mice with EAM into naive wild-type recipients reproduced the severity of the disease, demonstrating that CD69 exerts its function within the lymphocyte compartment. CONCLUSION: Our findings indicate that CD69 negatively regulates heart-specific Th17 responses, cardiac inflammation, and heart failure progression in EAM.


Asunto(s)
Enfermedades Autoinmunes/fisiopatología , Lectinas Tipo C/deficiencia , Miocarditis/fisiopatología , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/genética , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Cruzamientos Genéticos , Fibrosis Endomiocárdica/inmunología , Fibrosis Endomiocárdica/fisiopatología , Femenino , Citometría de Flujo , Humanos , Lectinas Tipo C/genética , Ganglios Linfáticos/citología , Ganglios Linfáticos/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Miocarditis/genética , Miocarditis/inmunología , Cadenas Pesadas de Miosina/inmunología , Fragmentos de Péptidos/inmunología , Índice de Severidad de la Enfermedad , Bazo/citología , Bazo/fisiología
14.
Proc Natl Acad Sci U S A ; 105(30): 10507-12, 2008 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-18641128

RESUMEN

Ras/ERK signaling plays an important role in T cell activation and development. We recently reported that endothelial nitric oxide synthase (eNOS)-derived NO regulates T cell receptor (TCR)-dependent ERK activation by a cGMP-independent mechanism. Here, we explore the mechanisms through which eNOS exerts this regulation. We have found that eNOS-derived NO positively regulates Ras/ERK activation in T cells stimulated with antigen on antigen-presenting cells (APCs). Intracellular activation of N-, H-, and K-Ras was monitored with fluorescent probes in T cells stably transfected with eNOS-GFP or its G2A point mutant, which is defective in activity and cellular localization. Using this system, we demonstrate that eNOS selectively activates N-Ras but not K-Ras on the Golgi complex of T cells engaged with APC, even though Ras isoforms are activated in response to NO from donors. We further show that activation of N-Ras involves eNOS-dependent S-nitrosylation on Cys(118), suggesting that upon TCR engagement, eNOS-derived NO directly activates N-Ras on the Golgi. Moreover, wild-type but not C118S N-Ras increased TCR-dependent apoptosis, suggesting that S-nitrosylation of Cys(118) contributes to activation-induced T cell death. Our data define a signaling mechanism for the regulation of the Ras/ERK pathway based on the eNOS-dependent differential activation of N-Ras and K-Ras at specific cell compartments.


Asunto(s)
Antígenos/química , Apoptosis , Regulación Enzimológica de la Expresión Génica , Aparato de Golgi/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Linfocitos T/inmunología , Proteínas ras/metabolismo , Antígenos CD28/química , Cisteína/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Modelos Biológicos , Óxido Nítrico/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-raf/metabolismo , Linfocitos T/metabolismo
15.
J Allergy Clin Immunol ; 126(2): 355-65, 365.e1-3, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20621339

RESUMEN

BACKGROUND: Allergic diseases have a major health care impact in industrialized countries. The development of these diseases is influenced by exposure to allergen and to immunological and genetic factors. However, the molecular mechanisms underlying the inflammatory response that triggers allergy are not well defined. OBJECTIVE: We have investigated the role of the leukocyte activation antigen CD69 in the regulation of two allergic diseases, asthma and contact dermatitis. METHODS: Analysis of two models of allergic diseases in CD69 knockout and wild-type mice: ovalbumin-induced allergic airway inflammation (BALB/c genetic background) and contact hypersensitivity to oxazolone (C57BL/6J genetic background). RESULTS: CD69 deficiency dramatically enhanced the inflammatory response in the ovalbumin-induced asthma model of antigen-induced airway allergy. CD69 knockout mice showed exacerbated pulmonary eosinophil recruitment, high vascular cell adhesion molecule 1 expression levels in lung vasculature, and enhanced T(H)2 and T(H)17 cytokines in the bronchoalveolar space and lung tissue. In the hapten-induced cutaneous contact hypersensitivity model, both CD69 deficiency and treatment with anti-CD69 mAb increased inflammation. Treatment with contact allergens induced enhanced T(H)1 and T(H)17 responses in CD69 deficient mice, and neutralizing anti-IL-17 antibodies reduced skin inflammation. In both experimental systems, adoptive transfer of lymph node cells from CD69 knockout mice increased the inflammatory response in recipient mice. CONCLUSION: These results demonstrate that the early activation receptor CD69 is an intrinsic modulator of immune allergic processes through the negative regulation of allergen-induced T-cell effector responses.


Asunto(s)
Asma/inmunología , Dermatitis Alérgica por Contacto/inmunología , Eosinófilos/inmunología , Células TH1/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Antígenos CD , Antígenos de Diferenciación de Linfocitos T , Asma/genética , Asma/patología , Dermatitis Alérgica por Contacto/genética , Dermatitis Alérgica por Contacto/patología , Modelos Animales de Enfermedad , Eosinófilos/patología , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/genética , Interleucina-17/inmunología , Lectinas Tipo C , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células TH1/patología , Células Th2/patología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología
16.
Nat Commun ; 12(1): 6230, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34711828

RESUMEN

T cells undergo rigorous selection in the thymus to ensure self-tolerance and prevent autoimmunity, with this process requiring innocuous self-antigens (Ags) to be presented to thymocytes. Self-Ags are either expressed by thymic stroma cells or transported to the thymus from the periphery by migratory dendritic cells (DCs); meanwhile, small blood-borne peptides can access the thymic parenchyma by diffusing across the vascular lining. Here we describe an additional pathway of thymic Ag acquisition that enables circulating antigenic macromolecules to access both murine and human thymi. This pathway depends on a subset of thymus-resident DCs, distinct from both parenchymal and circulating migratory DCs, that are positioned in immediate proximity to thymic microvessels where they extend cellular processes across the endothelial barrier into the blood stream. Transendothelial positioning of DCs depends on DC-expressed CX3CR1 and its endothelial ligand, CX3CL1, and disrupting this chemokine pathway prevents thymic acquisition of circulating proteins and compromises negative selection of Ag-reactive thymocytes. Thus, transendothelial DCs represent a mechanism by which the thymus can actively acquire blood-borne Ags to induce and maintain central tolerance.


Asunto(s)
Sangre/inmunología , Células Dendríticas/inmunología , Células Endoteliales/inmunología , Timocitos/inmunología , Timo/inmunología , Animales , Autoantígenos/inmunología , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Diferenciación Celular , Movimiento Celular , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/inmunología , Células Dendríticas/citología , Células Endoteliales/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Autotolerancia , Timocitos/citología , Timo/citología
17.
Blood ; 112(8): 3217-26, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18663148

RESUMEN

MT1-MMP plays a key role in endothelial function, as underscored by the angiogenic defects found in MT1-MMP deficient mice. We have studied the molecular interactions that underlie the functional regulation of MT1-MMP. At lateral endothelial cell junctions, MT1-MMP colocalizes with tetraspanin CD151 (Tspan 24) and its associated partner alpha3beta1 integrin. Biochemical and FRET analyses show that MT1-MMP, through its hemopexin domain, associates tightly with CD151, thus forming alpha3beta1 integrin/CD151/MT1-MMP ternary complexes. siRNA knockdown of HUVEC CD151 expression enhanced MT1-MMP-mediated activation of MMP2, and the same activation was seen in ex vivo lung endothelial cells isolated from CD151-deficient mice. However, analysis of collagen degradation in these experimental models revealed a diminished MT1-MMP enzymatic activity in confined areas around the cell periphery. CD151 knockdown affected both MT1-MMP subcellular localization and its inclusion into detergent-resistant membrane domains, and prevented biochemical association of the metalloproteinase with the integrin alpha3beta1. These data provide evidence for a novel regulatory role of tetraspanin microdomains on the collagenolytic activity of MT1-MMP and indicate that CD151 is a key regulator of MT1-MMP in endothelial homeostasis.


Asunto(s)
Antígenos CD/química , Células Endoteliales/citología , Regulación de la Expresión Génica , Metaloproteinasa 14 de la Matriz/metabolismo , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Activación Enzimática , Transferencia Resonante de Energía de Fluorescencia , Homeostasis , Humanos , Integrina alfa3beta1/metabolismo , Pulmón/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Tetraspanina 24
18.
Hepatology ; 48(4): 1044-53, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18802961

RESUMEN

UNLABELLED: Hepatocyte tight junctions (TJ) play key roles in characteristic liver functions, including bile formation and secretion. Infection by hepatitis C virus (HCV) may cause alterations of the liver architecture and disruption of the bile duct, which ultimately can lead to cholestasis. Herein, we employed the HCV replicon system to analyze the effect of HCV on TJ organization. TJ-associated proteins occludin, claudin-1, and Zonula Occludens protein-1 (ZO-1) disappeared from their normal localization at the border of adjacent cells in Huh7 clones harboring genomic but not subgenomic replicons expressing only the nonstructural proteins. Furthermore, cells containing genomic replicons showed a cytoplasmic accumulation of occludin in the endoplasmic reticulum (ER). TJ-associated function, measured as FITC-dextran paracellular permeability, of genomic replicon-containing cells, was also altered. Interestingly, clearance of the HCV replicon by interferon-alpha (IFN-alpha) treatment and by short hairpin RNA (shRNA) significantly restored the localization of TJ-associated proteins. Transient expression of all HCV structural proteins, but not core protein alone, altered the localization of TJ-associated proteins in Huh7 cells and in clones with subgenomic replicons. Confocal analysis showed that accumulation of occludin in the ER partially co-localized with HCV envelope glycoprotein E2. E2/occludin association was further confirmed by co-immunoprecipitation and pull-down assays. Additionally, using a cell culture model of HCV infection, we observed the cytoplasmic dot-like accumulation of occludin in infected Huh7 cells. CONCLUSION: We propose that HCV structural proteins, most likely those of the viral envelope, promote alterations of TJ-associated proteins, which may provide new insights for HCV-related pathogenesis.


Asunto(s)
Retículo Endoplásmico/metabolismo , Hepacivirus/metabolismo , Hepatocitos/metabolismo , Proteínas de la Membrana/metabolismo , Uniones Estrechas/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Antivirales/farmacología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Claudina-1 , Silenciador del Gen , Genoma Viral/genética , Hepacivirus/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Interferón-alfa/farmacología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Ocludina , Fosfoproteínas/metabolismo , Replicón/genética , Proteína de la Zonula Occludens-1
19.
J Cell Biol ; 157(7): 1233-45, 2002 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-12082081

RESUMEN

Ezrin, radixin, and moesin (ERM) regulate cortical morphogenesis and cell adhesion by connecting membrane adhesion receptors to the actin-based cytoskeleton. We have studied the interaction of moesin and ezrin with the vascular cell adhesion molecule (VCAM)-1 during leukocyte adhesion and transendothelial migration (TEM). VCAM-1 interacted directly with moesin and ezrin in vitro, and all of these molecules colocalized at the apical surface of endothelium. Dynamic assessment of this interaction in living cells showed that both VCAM-1 and moesin were involved in lymphoblast adhesion and spreading on the endothelium, whereas only moesin participated in TEM, following the same distribution pattern as ICAM-1. During leukocyte adhesion in static or under flow conditions, VCAM-1, ICAM-1, and activated moesin and ezrin clustered in an endothelial actin-rich docking structure that anchored and partially embraced the leukocyte containing other cytoskeletal components such as alpha-actinin, vinculin, and VASP. Phosphoinositides and the Rho/p160 ROCK pathway, which participate in the activation of ERM proteins, were involved in the generation and maintenance of the anchoring structure. These results provide the first characterization of an endothelial docking structure that plays a key role in the firm adhesion of leukocytes to the endothelium during inflammation.


Asunto(s)
Endotelio Vascular/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/metabolismo , Proteínas de Microfilamentos/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Actinas/metabolismo , Línea Celular , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Glutatión Transferasa/metabolismo , Humanos , Recién Nacido , Leucocitos/citología , Activación de Linfocitos , Fosfoproteínas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Tubulina (Proteína)/metabolismo , Venas Umbilicales/citología
20.
Mol Biol Cell ; 17(8): 3435-45, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16738306

RESUMEN

In this work, the role of HDAC6, a type II histone deacetylase with tubulin deacetylase activity, in lymphocyte polarity, motility, and transmigration was explored. HDAC6 was localized at dynamic subcellular structures as leading lamellipodia and the uropod in migrating T-cells. However, HDAC6 activity did not appear to be involved in the polarity of migrating lymphocytes. Overexpression of HDAC6 in freshly isolated lymphocytes and T-cell lines increased the lymphocyte migration mediated by chemokines and their transendothelial migration under shear flow. Accordingly, the knockdown of HDAC6 expression in T-cells diminished their chemotactic capability. Additional experiments with HDAC6 inhibitors (trichostatin, tubacin), other structural related molecules (niltubacin, MAZ-1391), and HDAC6 dead mutants showed that the deacetylase activity of HDAC6 was not involved in the modulatory effect of this molecule on cell migration. Our results indicate that HDAC6 has an important role in the chemotaxis of T-lymphocytes, which is independent of its tubulin deacetylase activity.


Asunto(s)
Quimiotaxis , Histona Desacetilasas/metabolismo , Linfocitos T/citología , Linfocitos T/enzimología , Acetilación , Anilidas/química , Anilidas/farmacología , Adhesión Celular/efectos de los fármacos , Inhibición de Migración Celular , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Expresión Génica , Silenciador del Gen , Histona Desacetilasa 6 , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/deficiencia , Humanos , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Activación de Linfocitos/inmunología , Proteínas Mutantes/metabolismo , Transporte de Proteínas , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Tubulina (Proteína)/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda