Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
bioRxiv ; 2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37790300

RESUMEN

The growth of antimicrobial resistance (AMR) has highlighted an urgent need to identify bacterial pathogenic functions that may be targets for clinical intervention. Although severe bacterial infections profoundly alter host metabolism, prior studies have largely ignored alterations in microbial metabolism in this context. Performing metabolomics on patient and mouse plasma samples, we identify elevated levels of bacterially-derived N-acetylputrescine during gram-negative bloodstream infections (BSI), with higher levels associated with worse clinical outcomes. We discover that SpeG is the bacterial enzyme responsible for acetylating putrescine and show that blocking its activity reduces bacterial proliferation and slows pathogenesis. Reduction of SpeG activity enhances bacterial membrane permeability and results in increased intracellular accumulation of antibiotics, allowing us to overcome AMR of clinical isolates both in culture and in vivo. This study highlights how studying pathogen metabolism in the natural context of infection can reveal new therapeutic strategies for addressing challenging infections.

2.
J Innate Immun ; 15(1): 765-781, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37797588

RESUMEN

Novel therapeutics are urgently needed to prevent opportunistic infections in immunocompromised individuals undergoing cancer treatments or other immune-suppressive therapies. Trained immunity is a promising strategy to reduce this burden of disease. We previously demonstrated that mesenchymal stromal cells (MSCs) preconditioned with a class A CpG oligodeoxynucleotide (CpG-ODN), a Toll-like receptor 9 (TLR9) agonist, can augment emergency granulopoiesis in a murine model of neutropenic sepsis. Here, we used a chimeric mouse model to demonstrate that MSCs secrete paracrine factors that act on lineage-negative c-kit+ hematopoietic stem cells (HSCs), leaving them "poised" to enhance emergency granulopoiesis months after transplantation. Chimeric mice developed from HSCs exposed to conditioned media from MSCs and CpG-ODN-preconditioned MSCs showed significantly higher bacterial clearance and increased neutrophil granulopoiesis following lung infection than control mice. By Cleavage Under Targets and Release Using Nuclease (CUT&RUN) chromatin sequencing, we identified that MSC-conditioned media leaves H3K4me3 histone marks in HSCs at genes involved in myelopoiesis and in signaling persistence by the mTOR pathway. Both soluble factors and extracellular vesicles from MSCs mediated these effects on HSCs and proteomic analysis by mass spectrometry revealed soluble calreticulin as a potential mediator. In summary, this study demonstrates that trained immunity can be mediated by paracrine factors from MSCs to induce neutrophil-trained immunity by reprogramming HSCs for long-lasting functional changes in neutrophil-mediated antimicrobial immunity.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Ratones , Animales , Neutrófilos , Medios de Cultivo Condicionados/metabolismo , Proteómica , Inmunidad Entrenada , Células Madre Hematopoyéticas , Células Madre Mesenquimatosas/metabolismo
3.
Front Immunol ; 13: 833100, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35634302

RESUMEN

CD4+ regulatory T cells (Tregs) activate and expand in response to different types of injuries, suggesting that they play a critical role in controlling the immune response to tissue and cell damage. This project used multi-dimensional profiling techniques to comprehensively characterize injury responsive Tregs in mice. We show that CD44high Tregs expand in response to injury and were highly suppressive when compared to CD44low Tregs. T cell receptor (TCR) repertoire analysis revealed that the CD44high Treg population undergo TCRαß clonal expansion as well as increased TCR CDR3 diversity. Bulk RNA sequencing and single-cell RNA sequencing with paired TCR clonotype analysis identified unique differences between CD44high and CD44low Tregs and specific upregulation of genes in Tregs with expanded TCR clonotypes. Gene ontology analysis for molecular function of RNA sequencing data identified chemokine receptors and cell division as the most enriched functional terms in CD44high Tregs versus CD44low Tregs. Mass cytometry (CyTOF) analysis of Tregs from injured and uninjured mice verified protein expression of these genes on CD44high Tregs, with injury-induced increases in Helios, Galectin-3 and PYCARD expression. Taken together, these data indicate that injury triggers the expansion of a highly suppressive CD44high Treg population that is transcriptionally and phenotypically distinct from CD44low Tregs suggesting that they actively participate in controlling immune responses to injury and tissue damage.


Asunto(s)
Receptores de Antígenos de Linfocitos T alfa-beta , Linfocitos T Reguladores , Animales , Proliferación Celular , Ratones
4.
Life Sci Alliance ; 5(5)2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35181635

RESUMEN

Lymphangioleiomyomatosis (LAM) is a rare progressive disease, characterized by mutations in the tuberous sclerosis complex genes (TSC1 or TSC2) and hyperactivation of mechanistic target of rapamycin complex 1 (mTORC1). Here, we report that E26 transformation-specific (ETS) variant transcription factor 2 (ETV2) is a critical regulator of Tsc2-deficient cell survival. ETV2 nuclear localization in Tsc2-deficient cells is mTORC1-independent and is enhanced by spleen tyrosine kinase (Syk) inhibition. In the nucleus, ETV2 transcriptionally regulates poly(ADP-ribose) polymerase 1 binding protein (PARPBP) mRNA and protein expression, partially reversing the observed down-regulation of PARPBP expression induced by mTORC1 blockade during treatment with both Syk and mTORC1 inhibitors. In addition, silencing Etv2 or Parpbp in Tsc2-deficient cells induced ER stress and increased cell death in vitro and in vivo. We also found ETV2 expression in human cells with loss of heterozygosity for TSC2, lending support to the translational relevance of our findings. In conclusion, we report a novel ETV2 signaling axis unique to Syk inhibition that promotes a cytocidal response in Tsc2-deficient cells and therefore maybe a potential alternative therapeutic target in LAM.


Asunto(s)
Linfangioleiomiomatosis , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Proteínas de Unión al ADN/genética , Estrés del Retículo Endoplásmico , Humanos , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Factores de Transcripción/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
5.
Blood Adv ; 6(7): 2001-2013, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-34991159

RESUMEN

Infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) engages the inflammasome in monocytes and macrophages and leads to the cytokine storm in COVID-19. Neutrophils, the most abundant leukocytes, release neutrophil extracellular traps (NETs), which have been implicated in the pathogenesis of COVID-19. Our recent study shows that activation of the NLRP3 inflammasome is important for NET release in sterile inflammation. However, the role of neutrophil inflammasome formation in human disease is unknown. We hypothesized that SARS-CoV-2 infection may induce inflammasome activation in neutrophils. We also aimed to assess the localization of inflammasome formation (ie, apoptosis-associated speck-like protein containing a CARD [ASC] speck assembly) and timing relative to NETosis in stimulated neutrophils by real-time video microscopy. Neutrophils isolated from severe COVID-19 patients demonstrated that ∼2% of neutrophils in both the peripheral blood and tracheal aspirates presented ASC speck. ASC speck was observed in neutrophils with an intact poly-lobulated nucleus, suggesting early formation during neutrophil activation. Additionally, 40% of nuclei were positive for citrullinated histone H3, and there was a significant correlation between speck formation and nuclear histone citrullination. Time-lapse microscopy in lipopolysaccharide -stimulated neutrophils from fluorescent ASC reporter mice showed that ASC speck formed transiently and at the microtubule organizing center long before NET release. Our study shows that ASC speck is present in neutrophils from COVID-19 patients with respiratory failure and that it forms early in NETosis. Our findings suggest that inhibition of neutrophil inflammasomes may be beneficial in COVID-19.


Asunto(s)
COVID-19 , Trampas Extracelulares , Animales , Trampas Extracelulares/metabolismo , Humanos , Inflamasomas/metabolismo , Ratones , Neutrófilos/metabolismo , SARS-CoV-2
6.
FEBS J ; 289(2): 417-435, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34355516

RESUMEN

Sepsis is a life-threatening process related to a dysregulated host response to an underlying infection, which results in organ dysfunction and poor outcomes. Therapeutic strategies using mesenchymal stromal cells (MSCs) are under investigation for sepsis, with efforts to improve cellular utility. Syndecan (SDC) proteins are transmembrane proteoglycans involved with cellular signaling events including tissue repair and modulating inflammation. Bone marrow-derived human MSCs express syndecan-2 (SDC2) at a level higher than other SDC family members; thus, we explored SDC2 in MSC function. Administration of human MSCs silenced for SDC2 in experimental sepsis resulted in decreased bacterial clearance, and increased tissue injury and mortality compared with wild-type MSCs. These findings were associated with a loss of resolution of inflammation in the peritoneal cavity, and higher levels of proinflammatory mediators in organs. MSCs silenced for SDC2 had a decreased ability to promote phagocytosis of apoptotic neutrophils by macrophages in the peritoneum, and also a diminished capability to convert macrophages from a proinflammatory to a proresolution phenotype via cellular or paracrine actions. Extracellular vesicles are a paracrine effector of MSCs that may contribute to resolution of inflammation, and their production was dramatically reduced in SDC2-silenced human MSCs. Collectively, these data demonstrate the importance of SDC2 for cellular and paracrine function of human MSCs during sepsis.


Asunto(s)
Vesículas Extracelulares/genética , Inflamación/genética , Sepsis/genética , Sindecano-2/genética , Animales , Polaridad Celular/genética , Polaridad Celular/inmunología , Modelos Animales de Enfermedad , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/microbiología , Regulación del Desarrollo de la Expresión Génica/genética , Silenciador del Gen , Humanos , Inmunidad/genética , Inflamación/microbiología , Inflamación/patología , Inflamación/terapia , Macrófagos/inmunología , Macrófagos/microbiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Neutrófilos/inmunología , Neutrófilos/microbiología , Comunicación Paracrina/genética , Fagocitosis/genética , Sepsis/microbiología , Sepsis/patología , Sepsis/terapia
7.
JCI Insight ; 6(22)2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34665782

RESUMEN

Lung allograft rejection results in the accumulation of low-molecular weight hyaluronic acid (LMW-HA), which further propagates inflammation and tissue injury. We have previously shown that therapeutic lymphangiogenesis in a murine model of lung allograft rejection reduced tissue LMW-HA and was associated with improved transplant outcomes. Herein, we investigated the use of 4-Methylumbelliferone (4MU), a known inhibitor of HA synthesis, to alleviate acute allograft rejection in a murine model of lung transplantation. We found that treating mice with 4MU from days 20 to 30 after transplant was sufficient to significantly improve outcomes, characterized by a reduction in T cell-mediated lung inflammation and LMW-HA content and in improved pathology scores. In vitro, 4MU directly attenuated activation, proliferation, and differentiation of naive CD4+ T cells into Th1 cells. As 4MU has already been demonstrated to be safe for human use, we believe examining 4MU for the treatment of acute lung allograft rejection may be of clinical significance.


Asunto(s)
Rechazo de Injerto/terapia , Ácido Hialurónico/efectos adversos , Trasplante de Pulmón/efectos adversos , Aloinjertos , Animales , Humanos , Trasplante de Pulmón/métodos , Ratones
8.
Respir Res ; 22(1): 277, 2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702264

RESUMEN

Lymphangioleiomyomatosis (LAM) is a progressive cystic lung disease with mortality driven primarily by respiratory failure. Patients with LAM frequently have respiratory infections, suggestive of a dysregulated microbiome. Here we demonstrate that end-stage LAM patients have a distinct microbiome signature compared to patients with end-stage chronic obstructive pulmonary disease.


Asunto(s)
Pulmón/microbiología , Linfangioleiomiomatosis/microbiología , Microbiota , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Infecciones del Sistema Respiratorio/microbiología , Progresión de la Enfermedad , Disbiosis , Humanos , Enfermedades Pulmonares Fúngicas/diagnóstico , Enfermedades Pulmonares Fúngicas/microbiología , Linfangioleiomiomatosis/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Infecciones del Sistema Respiratorio/diagnóstico , Ribotipificación
9.
Stem Cell Res Ther ; 12(1): 487, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34461993

RESUMEN

BACKGROUND: Acute lung injury (ALI) is a common lung disorder that affects millions of people every year. The infiltration of inflammatory cells into the lungs and death of the alveolar epithelial cells are key factors to trigger a pathological cascade. Trophoblast stem cells (TSCs) are immune privileged, and demonstrate the capability of self-renewal and multipotency with differentiation into three germ layers. We hypothesized that intratracheal transplantation of TSCs may alleviate ALI. METHODS: ALI was induced by intratracheal delivery of bleomycin (BLM) in mice. After exposure to BLM, pre-labeled TSCs or fibroblasts (FBs) were intratracheally administered into the lungs. Analyses of the lungs were performed for inflammatory infiltrates, cell apoptosis, and engraftment of TSCs. Pro-inflammatory cytokines/chemokines of lung tissue and in bronchoalveolar lavage fluid (BALF) were also assessed. RESULTS: The lungs displayed a reduction in cellularity, with decreased CD45+ cells, and less thickening of the alveolar walls in ALI mice that received TSCs compared with ALI mice receiving PBS or FBs. TSCs decreased infiltration of neutrophils and macrophages, and the expression of interleukin (IL) 6, monocyte chemoattractant protein-1 (MCP-1) and keratinocyte-derived chemokine (KC) in the injured lungs. The levels of inflammatory cytokines in BALF, particularly IL-6, were decreased in ALI mice receiving TSCs, compared to ALI mice that received PBS or FBs. TSCs also significantly reduced BLM-induced apoptosis of alveolar epithelial cells in vitro and in vivo. Transplanted TSCs integrated into the alveolar walls and expressed aquaporin 5 and prosurfactant protein C, markers for alveolar epithelial type I and II cells, respectively. CONCLUSION: Intratracheal transplantation of TSCs into the lungs of mice after acute exposure to BLM reduced pulmonary inflammation and cell death. Furthermore, TSCs engrafted into the alveolar walls to form alveolar epithelial type I and II cells. These data support the use of TSCs for the treatment of ALI.


Asunto(s)
Lesión Pulmonar Aguda , Trofoblastos , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/terapia , Células Epiteliales Alveolares , Animales , Líquido del Lavado Bronquioalveolar , Lipopolisacáridos , Pulmón , Ratones , Ratones Endogámicos C57BL , Células Madre
10.
JCI Insight ; 6(18)2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34403374

RESUMEN

Oligoarticular juvenile idiopathic arthritis (oligo JIA) is the most common form of chronic inflammatory arthritis in children, yet the cause of this disease remains unknown. To understand immune responses in oligo JIA, we immunophenotyped synovial fluid T cells with flow cytometry, bulk RNA-Seq, single-cell RNA-Seq (scRNA-Seq), DNA methylation studies, and Treg suppression assays. In synovial fluid, CD4+, CD8+, and γδ T cells expressed Th1-related markers, whereas Th17 cells were not enriched. Th1 skewing was prominent in CD4+ T cells, including Tregs, and was associated with severe disease. Transcriptomic studies confirmed a Th1 signature in CD4+ T cells from synovial fluid. The regulatory gene expression signature was preserved in Tregs, even those exhibiting Th1 polarization. These Th1-like Tregs maintained Treg-specific methylation patterns and suppressive function, supporting the stability of this Treg population in the joint. Although synovial fluid CD4+ T cells displayed an overall Th1 phenotype, scRNA-Seq uncovered heterogeneous effector and regulatory subpopulations, including IFN-induced Tregs, peripheral helper T cells, and cytotoxic CD4+ T cells. In conclusion, oligo JIA is characterized by Th1 polarization that encompasses Tregs but does not compromise their regulatory identity. Targeting Th1-driven inflammation and augmenting Treg function may represent important therapeutic approaches in oligo JIA.


Asunto(s)
Artritis Juvenil/inmunología , Polaridad Celular , Líquido Sinovial/inmunología , Linfocitos T/fisiología , Adolescente , Artritis Juvenil/genética , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Estudios de Casos y Controles , Niño , Preescolar , Metilación de ADN , Femenino , Humanos , Inmunofenotipificación , Lactante , Linfocitos Intraepiteliales/fisiología , Masculino , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Linfocitos T Reguladores/fisiología , Células TH1/fisiología , Transcriptoma
11.
J Leukoc Biol ; 110(4): 711-722, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33438259

RESUMEN

High mobility group (HMG)A proteins are nonhistone chromatin proteins that bind to the minor groove of DNA, interact with transcriptional machinery, and facilitate DNA-directed nuclear processes. HMGA1 has been shown to regulate genes involved with systemic inflammatory processes. We hypothesized that HMGA1 is important in the function of mesenchymal stromal cells (MSCs), which are known to modulate inflammatory responses due to sepsis. To study this process, we harvested MSCs from transgenic (Tg) mice expressing a dominant-negative (dn) form of HMGA1 in mesenchymal cells. MSCs harvested from Tg mice contained the dnHMGA1 transgene, and transgene expression did not change endogenous HMGA1 levels. Immunophenotyping of the cells, along with trilineage differentiation revealed no striking differences between Tg and wild-type (WT) MSCs. However, Tg MSCs growth was decreased compared with WT MSCs, although Tg MSCs were more resistant to oxidative stress-induced death and expressed less IL-6. Tg MSCs administered after the onset of Escherichia coli-induced sepsis maintained their ability to improve survival when given in a single dose, in contrast with WT MSCs. This survival benefit of Tg MSCs was associated with less tissue cell death, and also a reduction in tissue neutrophil infiltration and expression of neutrophil chemokines. Finally, Tg MSCs promoted bacterial clearance and enhanced neutrophil phagocytosis, in part through their increased expression of stromal cell-derived factor-1 compared with WT MSCs. Taken together, these data demonstrate that expression of dnHMGA1 in MSCs provides a functional advantage of the cells when administered during bacterial sepsis.


Asunto(s)
Genes Dominantes , Proteína HMGA1a/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Sepsis/patología , Sepsis/terapia , Transgenes , Adipocitos/citología , Animales , Muerte Celular , Proliferación Celular , Supervivencia Celular , Quimiocina CXCL12/biosíntesis , Escherichia coli/fisiología , Proteína HMGA1a/metabolismo , Inflamación/patología , Interleucina-6/biosíntesis , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Infiltración Neutrófila , Neutrófilos/metabolismo , Estrés Oxidativo , Fagocitosis , Sepsis/microbiología
12.
Blood Adv ; 4(19): 4965-4979, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33049055

RESUMEN

Patients with immune deficiencies from cancers and associated treatments represent a growing population within the intensive care unit with increased risk of morbidity and mortality from sepsis. Mesenchymal stromal cells (MSCs) are an integral part of the hematopoietic niche and express toll-like receptors, making them candidate cells to sense and translate pathogenic signals into an innate immune response. In this study, we demonstrate that MSCs administered therapeutically in a murine model of radiation-associated neutropenia have dual actions to confer a survival benefit in Pseudomonas aeruginosa pneumo-sepsis that is not from improved bacterial clearance. First, MSCs augment the neutrophil response to infection, an effect that is enhanced when MSCs are preconditioned with CpG oligodeoxynucleotide, a toll-like receptor 9 agonist. Using cytometry by time of flight, we identified proliferating neutrophils (Ly6GlowKi-67+) as the main expanded cell population within the bone marrow. Further analysis revealed that CpG-MSCs expand a lineage restricted progenitor population (Lin-Sca1+C-kit+CD150-CD48+) in the bone marrow, which corresponded to a doubling in the myeloid proliferation and differentiation potential in response to infection compared with control. Despite increased neutrophils, no reduction in organ bacterial count was observed between experimental groups. However, the second effect exerted by CpG-MSCs is to attenuate organ damage, particularly in the lungs. Neutrophils obtained from irradiated mice and cocultured with CpG-MSCs had decreased neutrophil extracellular trap formation, which was associated with decreased citrullinated H3 staining in the lungs of mice given CpG-MSCs in vivo. Thus, this preclinical study provides evidence for the therapeutic potential of MSCs in neutropenic sepsis.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Neutropenia , Sepsis , Animales , Hematopoyesis , Humanos , Ratones , Neutropenia/terapia , Sepsis/terapia
13.
Adv Pharm Bull ; 10(3): 418-422, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32665900

RESUMEN

Purpose: Complete recycling of the crop residues of sugarcane in the Philippines remains to be achieved. This study purposed to derive microcrystalline cellulose (MCC) from sugarcane leaves and test its disintegrating properties in tablet formulation. Methods: Saccharum officinarum L. (sugarcane) leaves were used to prepare MCC powder. According to the conventional method, the preparation of cellulose powder requires heating the raw material with acid and alkali followed by washing, bleaching, and sieving. Hydrolysis of the bleached product was carried out using hydrochloric acid to obtain MCC powder, and the physicochemical properties of the produced MCC powder were studied including its organoleptic properties, pH value, %loss on drying, %water soluble substances and Fouriertransform infrared (FTIR) spectrum. Results: The resulting powder was evaluated for its disintegrating property in the preparation of blank tablets, which were compared to tablets prepared using commercially available MCC. MCC powder derived from sugarcane leaves had properties at par with commercially available MCC and was in conformance with National Formulary (NF) specifications. Conclusion: Disintegrating properties were also significantly better than the commercially available MCC.

14.
Crit Care Med ; 48(5): e409-e417, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32167490

RESUMEN

OBJECTIVES: Sepsis results in organ dysfunction caused by a dysregulated host response, in part related to the immune response of a severe infection. Mesenchymal stromal cells are known to modulate the immune response, and expression of stromal cell-derived factor-1 regulates mobilization of neutrophils from the bone marrow. We are investigating the importance of stromal cell-derived factor-1 in mesenchymal stromal cells and its role in promoting neutrophil function after the onset of cecal ligation and puncture-induced sepsis. Stromal cell-derived factor-1 expression was silenced in mesenchymal stromal cells, compared with the control scrambled construct mesenchymal stromal cells. DESIGN: Animal study and cell culture. SETTING: Laboratory investigation. SUBJECTS: BALB/c mice. INTERVENTIONS: Polymicrobial sepsis was induced by cecal ligation and puncture. shSCR mesenchymal stromal cells and shSDF-1 mesenchymal stromal cells were delivered by tail vein injections to septic mice. The mice were assessed for survival, bacterial clearance, and the inflammatory response during sepsis in each of the groups. Mesenchymal stromal cells were also assessed for their ability to promote bacterial phagocytosis by neutrophils. MEASUREMENTS AND MAIN RESULTS: Injection of shSCR mesenchymal stromal cells after the onset of sepsis led to an increase in mouse survival (70%) at 7 days, whereas survival of mice receiving shSDF-1 mesenchymal stromal cells was significantly diminished (33%). The loss of survival benefit in mice receiving shSDF-1 mesenchymal stromal cells was associated with less efficient bacterial clearance compared with shSCR mesenchymal stromal cells. Although shSCR mesenchymal stromal cells, or their conditioned medium, were able to increase neutrophil phagocytosis of bacteria, this effect was significantly blunted with shSDF-1 mesenchymal stromal cells. Assessment of peritoneal inflammation revealed that neutrophils were significantly increased and more immature in septic mice receiving shSDF-1 mesenchymal stromal cells. This response was associated with hypocellularity and increased neutrophil death in the bone marrow of mice receiving shSDF-1 mesenchymal stromal cells. CONCLUSIONS: Expression of stromal cell-derived factor-1 in mesenchymal stromal cells enhances neutrophil function with increased phagocytosis, more efficient clearance of bacteria, and bone marrow protection from depletion of cellular reserves during sepsis.


Asunto(s)
Quimiocina CXCL12/farmacología , Células Madre Mesenquimatosas/fisiología , Sepsis/terapia , Animales , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Neutrófilos/metabolismo , Fagocitosis/efectos de los fármacos , Sepsis/mortalidad
15.
Chest ; 155(5): e117-e121, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31060706

RESUMEN

Patients with common variable immunodeficiency (CVID) can develop granulomatous-lymphocytic interstitial lung disease (GLILD), which is associated with increased morbidity and mortality. Treating GLILD is a significant challenge because it is rare and can be pathologically heterogeneous. Here we describe two cases of patients with CVID-associated GLILD with biopsies demonstrating loosely organized tertiary lymphoid structures (TLSs). Based on the pivotal role that B cells play in TLS initiation and maintenance, we hypothesized that using rituximab monotherapy for B-cell depletion alone would be sufficient for the disruption of the pathologic process underlying GLILD. These two cases demonstrate that adapting a strategy of B cell depletion monotherapy may be effective in TLS-associated conditions such as GLILD.


Asunto(s)
Inmunodeficiencia Variable Común/complicaciones , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Enfermedades Pulmonares Intersticiales/patología , Rituximab/uso terapéutico , Nódulo Pulmonar Solitario/tratamiento farmacológico , Nódulo Pulmonar Solitario/patología , Adulto , Antineoplásicos Inmunológicos/uso terapéutico , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Biopsia con Aguja , Inmunodeficiencia Variable Común/diagnóstico , Inmunodeficiencia Variable Común/inmunología , Femenino , Estudios de Seguimiento , Granuloma/inmunología , Granuloma/patología , Humanos , Inmunohistoquímica , Enfermedades Pulmonares Intersticiales/diagnóstico por imagen , Enfermedades Pulmonares Intersticiales/etiología , Masculino , Neumonectomía/métodos , Muestreo , Nódulo Pulmonar Solitario/diagnóstico por imagen , Nódulo Pulmonar Solitario/cirugía , Tomografía Computarizada por Rayos X/métodos , Resultado del Tratamiento
16.
Ann Am Thorac Soc ; 16(9): 1131-1137, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31026404

RESUMEN

Rationale: There is significant variation in approach to pre-lung transplant donor-specific antibodies (DSA), with some centers declining to cross any DSA. We implemented a protocol for transplantation for candidates with pretransplant DSA so long as a prospective complement-dependent cytotoxicity crossmatch was negative, regardless of number, specificity, class, or mean fluorescence intensity.Objectives: To compare post-transplant outcomes including overall survival, chronic lung allograft dysfunction-free survival, antibody-mediated rejection, and acute cellular rejection in lung transplant recipients where pretransplant DSA was and was not present.Methods: This was a single-center retrospective cohort study. For recipients with pretransplant DSA, if the prospective complement-dependent cytotoxicity crossmatch was negative, the donor offer was accepted and plasmapheresis was performed within 24 hours of transplantation and continued until retrospective crossmatch results returned. Immunosuppression and post-transplant management were not otherwise modified.Results: Of the 203 included recipients, 18 (8.9%) had pretransplant DSA. The median DSA mean fluorescence intensity was 4,000 (interquartile range, 2,975-5,625; total range, 2,100-17,000). The median number of DSA present per patient was one (interquartile range, 1-2). The presence of pretransplant DSA was not associated with increased mortality (hazard ratio, 1.2; 95% confidence interval [CI], 0.4-3.4) or decreased chronic lung allograft dysfunction-free survival (hazard ratio, 1.1; 95% CI, 0.6-2.1). Recipients with pretransplant DSA were more likely to require prolonged mechanical ventilation (adjusted odds ratio, 7.0; 95% CI, 2.3-21.6) and to have antibody-mediated rejection requiring treatment (adjusted odds ratio, 7.5; 95% CI, 1.0-55.8).Conclusions: A protocol of accepting donor offers for lung transplant candidates with preformed, complement-dependent cytotoxicity crossmatch-negative DSA is associated with increased need for prolonged mechanical ventilation and antibody-mediated rejection without affecting short-term overall or chronic lung allograft dysfunction-free survival.


Asunto(s)
Antígenos HLA/inmunología , Isoanticuerpos/inmunología , Trasplante de Pulmón/efectos adversos , Donantes de Tejidos , Anciano , Boston , Femenino , Rechazo de Injerto/epidemiología , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Prueba de Histocompatibilidad , Humanos , Trasplante de Pulmón/mortalidad , Masculino , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Resultado del Tratamiento
20.
Nat Med ; 20(5): 493-502, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24784232

RESUMEN

Interferon-α (IFN-α) is essential for antiviral immunity, but in the absence of matrix metalloproteinase-12 (MMP-12) or IκBα (encoded by NFKBIA) we show that IFN-α is retained in the cytosol of virus-infected cells and is not secreted. Our findings suggest that activated IκBα mediates the export of IFN-α from virus-infected cells and that the inability of cells in Mmp12(-/-) but not wild-type mice to express IκBα and thus export IFN-α makes coxsackievirus type B3 infection lethal and renders respiratory syncytial virus more pathogenic. We show here that after macrophage secretion, MMP-12 is transported into virus-infected cells. In HeLa cells MMP-12 is also translocated to the nucleus, where it binds to the NFKBIA promoter, driving transcription. We also identified dual-regulated substrates that are repressed both by MMP-12 binding to the substrate's gene exons and by MMP-12-mediated cleavage of the substrate protein itself. Whereas intracellular MMP-12 mediates NFKBIA transcription, leading to IFN-α secretion and host protection, extracellular MMP-12 cleaves off the IFN-α receptor 2 binding site of systemic IFN-α, preventing an unchecked immune response. Consistent with an unexpected role for MMP-12 in clearing systemic IFN-α, treatment of coxsackievirus type B3-infected wild-type mice with a membrane-impermeable MMP-12 inhibitor elevates systemic IFN-α levels and reduces viral replication in pancreas while sparing intracellular MMP-12. These findings suggest that inhibiting extracellular MMP-12 could be a new avenue for the development of antiviral treatments.


Asunto(s)
Núcleo Celular/genética , Inmunidad/genética , Interferón-alfa/genética , Metaloproteinasa 12 de la Matriz/genética , Animales , Sitios de Unión , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Citosol/metabolismo , Citosol/virología , Células HeLa , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Interferón-alfa/inmunología , Interferón-alfa/metabolismo , Metaloproteinasa 12 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Inhibidor NF-kappaB alfa , Páncreas/inmunología , Páncreas/virología , Virus del Sarcoma de Rous/genética , Virus del Sarcoma de Rous/patogenicidad , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...