Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(19): e2317307121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38683990

RESUMEN

Directing antibodies to a particular epitope among many possible on a target protein is a significant challenge. Here, we present a simple and general method for epitope-directed selection (EDS) using a differential phage selection strategy. This involves engineering the protein of interest (POI) with the epitope of interest (EOI) mutated using a systematic bioinformatics algorithm to guide the local design of an EOI decoy variant. Using several alternating rounds of negative selection with the EOI decoy variant followed by positive selection on the wild-type POI, we were able to identify highly specific and potent antibodies to five different EOI antigens that bind and functionally block known sites of proteolysis. Among these, we developed highly specific antibodies that target the proteolytic site on the CUB domain containing protein 1 (CDCP1) to prevent its proteolysis allowing us to study the cellular maturation of this event that triggers malignancy. We generated antibodies that recognize the junction between the pro- and catalytic domains for three different matrix metalloproteases (MMPs), MMP1, MMP3, and MMP9, that selectively block activation of each of these enzymes and impair cell migration. We targeted a proteolytic epitope on the cell surface receptor, EPH Receptor A2 (EphA2), that is known to transform it from a tumor suppressor to an oncoprotein. We believe that the EDS method greatly facilitates the generation of antibodies to specific EOIs on a wide range of proteins and enzymes for broad therapeutic and diagnostic applications.


Asunto(s)
Epítopos , Epítopos/inmunología , Humanos , Proteolisis , Unión Proteica , Ingeniería de Proteínas/métodos , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/inmunología , Anticuerpos/inmunología , Biblioteca de Péptidos
2.
J Interferon Cytokine Res ; 42(8): 352-368, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35647937

RESUMEN

The costs of coronavirus disease 2019 (COVID-19) are devastating. With millions of deaths worldwide, specific serological biomarkers, antiviral agents, and novel therapies are urgently required to reduce the disease burden. For these purposes, a profound understanding of the pathobiology of COVID-19 is mandatory. Notably, the study of immunity against other respiratory infections has generated reference knowledge to comprehend the paradox of the COVID-19 pathogenesis. Past studies point to a complex interplay between cytokines and other factors mediating wound healing and extracellular matrix (ECM) remodeling that results in exacerbated inflammation, tissue injury, severe manifestations, and a sequela of respiratory infections. This review provides an overview of the immunological process elicited after severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. Also, we analyzed available data about the participation of matrix metalloproteinases (MMPs) and transforming growth factor-beta (TGF-ß) in immune responses of the lungs. Furthermore, we discuss their possible implications in severe COVID-19 and sequela, including pulmonary fibrosis, and remark on the potential of these molecules as biomarkers for diagnosis, prognosis, and treatment of convalescent COVID-19 patients. Our review provides a theoretical framework for future research aimed to discover molecular hallmarks that, combined with clinical features, could serve as therapeutic targets and reliable biomarkers of the different clinical forms of COVID-19, including convalescence.


Asunto(s)
COVID-19 , Metaloproteinasas de la Matriz , Factor de Crecimiento Transformador beta , Biomarcadores , COVID-19/inmunología , Costo de Enfermedad , Humanos , Metaloproteinasas de la Matriz/inmunología , SARS-CoV-2 , Factor de Crecimiento Transformador beta/inmunología
3.
Viruses ; 13(8)2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34452454

RESUMEN

Coxsackievirus A2 (CVA2) has emerged as an active pathogen that has been implicated in hand, foot, and mouth disease (HFMD) and herpangina outbreaks worldwide. It has been reported that severe cases with CVA2 infection develop into heart injury, which may be one of the causes of death. However, the mechanisms of CVA2-induced heart injury have not been well understood. In this study, we used a neonatal mouse model of CVA2 to investigate the possible mechanisms of heart injury. We detected CVA2 replication and apoptosis in heart tissues from infected mice. The activity of total aspartate transaminase (AST) and lactate dehydrogenase (LDH) was notably increased in heart tissues from infected mice. CVA2 infection also led to the disruption of cell-matrix interactions in heart tissues, including the increases of matrix metalloproteinase (MMP)3, MMP8, MMP9, connective tissue growth factor (CTGF) and tissue inhibitors of metalloproteinases (TIMP)4. Infiltrating leukocytes (CD45+ and CD11b+ cells) were observed in heart tissues of infected mice. Correspondingly, the expression levels of inflammatory cytokines in tissue lysates of hearts, including tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1ß), IL6 and monocyte chemoattractant protein-1 (MCP-1) were significantly elevated in CVA2 infected mice. Inflammatory signal pathways in heart tissues, including phosphatidylinositol 3-kinase (PI3K)-AKT, mitogen-activated protein kinases (MAPK) and nuclear factor kappa B (NF-κB), were also activated after infection. In summary, CVA2 infection leads to heart injury in a neonatal mouse model, which might be related to viral replication, increased expression levels of MMP-related enzymes and excessive inflammatory responses.


Asunto(s)
Infecciones por Coxsackievirus/complicaciones , Enterovirus/patogenicidad , Lesiones Cardíacas/virología , Corazón/virología , Inflamación/virología , Animales , Animales Recién Nacidos , Apoptosis , Citocinas/clasificación , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Enterovirus/clasificación , Inflamación/inmunología , Metaloproteinasas de la Matriz/clasificación , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/inmunología , Ratones , Ratones Endogámicos BALB C , Transducción de Señal
4.
J Med Virol ; 93(8): 4629-4637, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33634515

RESUMEN

Globally, the burden due to dengue infection is increasing with a recent estimate of 96 million progressing to the disease every year. Dengue pathogenesis and the factors influencing it are not completely known. It is now widely speculated that there is an important role of matrix metalloproteinases (MMPs) in the initiation and progression of dengue pathogenesis; however, their exact roles are not fully understood. Overactivation of matrix metalloproteinases may contribute to the severity of dengue pathogenesis. Cytokines and various other mediators of inflammation interact with the vascular endothelium and matrix metalloproteinases may be one of the components among them. Extensive plasma leakage into tissue spaces may result in a shock. It is evident in the literature that MMP2 and MMP9 increase in dengue patients is correlated with the severity of the disease; however, the underlying mechanism is still unknown. Activation of innate cells and adaptive immune cells which include, B and T cells, macrophages or monocytes and dendritic cells also contribute to the dengue pathology. Newer therapeutic strategies include microRNAs, such as miR-134 (targets MMP3 and MMP1) and MicroRNA-320d, (targets MMP/TIMP proteolytic system). The use of antibodies-based therapeutics like (Andecaliximab; anti-matrix metalloproteinase-9 antibody) is also suggested against MMPs in dengue. In this review, we summarize some recent developments associated with the involvement of immune cells and their mediators associated with the matrix metalloproteinases mediated dengue pathogenesis. We highlight that, there is still very little knowledge about the MMPs in dengue pathogenesis which needs attention and extensive investigations.


Asunto(s)
Citocinas/inmunología , Virus del Dengue/inmunología , Dengue/inmunología , Dengue/terapia , Metaloproteinasas de la Matriz/inmunología , Dengue/enzimología , Dengue/patología , Humanos , Metaloproteinasa 2 de la Matriz/inmunología , Metaloproteinasa 9 de la Matriz/inmunología , Índice de Severidad de la Enfermedad
5.
Front Immunol ; 12: 788976, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095865

RESUMEN

Tuberculosis (TB) remains one of the leading infectious killers in the world, infecting approximately a quarter of the world's population with the causative organism Mycobacterium tuberculosis (M. tb). Central nervous system tuberculosis (CNS-TB) is the most severe form of TB, with high mortality and residual neurological sequelae even with effective TB treatment. In CNS-TB, recruited neutrophils infiltrate into the brain to carry out its antimicrobial functions of degranulation, phagocytosis and NETosis. However, neutrophils also mediate inflammation, tissue destruction and immunopathology in the CNS. Neutrophils release key mediators including matrix metalloproteinase (MMPs) which degrade brain extracellular matrix (ECM), tumor necrosis factor (TNF)-α which may drive inflammation, reactive oxygen species (ROS) that drive cellular necrosis and neutrophil extracellular traps (NETs), interacting with platelets to form thrombi that may lead to ischemic stroke. Host-directed therapies (HDTs) targeting these key mediators are potentially exciting, but currently remain of unproven effectiveness. This article reviews the key role of neutrophils and neutrophil-derived mediators in driving CNS-TB immunopathology.


Asunto(s)
Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Neutrófilos/inmunología , Tuberculosis/inmunología , Tuberculosis/metabolismo , Animales , Sistema Nervioso Central/microbiología , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Matriz Extracelular/microbiología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Metaloproteinasas de la Matriz/inmunología , Mycobacterium tuberculosis/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Tuberculosis/microbiología
6.
Int Immunol ; 32(1): 57-68, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31633754

RESUMEN

Increased T helper (Th)1/Th17 immune responses are a hallmark of Crohn's disease (CD) immunopathogenesis. CD90+ (myo-)fibroblasts (MFs) are abundant cells in the normal (N) intestinal mucosa contributing to mucosal tolerance via suppression of Th1 cell activity through cell surface membrane-bound PD-L1 (mPD-L1). CD-MFs have a decreased level of mPD-L1. Consequently, mPD-L1-mediated suppression of Th1 cells by CD-MFs is decreased, yet the mechanism responsible for the reduction in mPDL-1 is unknown. Increased expression of matrix metalloproteinases (MMPs) has been reported in CD. Herein we observed that when compared to N- and ulcerative colitis (UC)-MFs, CD-MFs increase in LPS-inducible levels of MMP-7 and -9 with a significant increase in both basal and inducible MMP-10. A similar pattern of MMP expression was observed in the CD-inflamed mucosa. Treatment of N-MFs with a combination of recombinant human MMP-7, -9 and -10 significantly decreased mPD-L1. In contrast, inhibition of MMP activity with MMP inhibitors or anti-MMP-10 neutralizing antibodies restores mPD-L1 on CD-MFs. CD-MFs demonstrated reduced capacity to suppress Th1 and Th17 responses from activated CD4+ T cells. By contrast, supplementation of the CD-MF:T-cell co-cultures with MMP inhibitors or anti-MMP neutralizing antibodies restored the CD-MF-mediated suppression. Our data suggest that (i) increased MMP-10 expression by CD-MFs and concomitant cleavage of PD-L1 from the surface of CD-MFs are likely to be one of the factors contributing to the decrease of mPD-L1-mediated suppression of Th1/Th17 cells in CD; and (ii) MMPs are likely to have a significant role in the intestinal mucosal immune responses.


Asunto(s)
Antígeno B7-H1/metabolismo , Membrana Celular/metabolismo , Enfermedad de Crohn/metabolismo , Fibroblastos/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Antígenos Thy-1/metabolismo , Antígeno B7-H1/inmunología , Membrana Celular/inmunología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Femenino , Fibroblastos/inmunología , Fibroblastos/patología , Humanos , Metaloproteinasas de la Matriz/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Antígenos Thy-1/inmunología
7.
J Tissue Viability ; 29(1): 42-47, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31866230

RESUMEN

BACKGROUND: The SIS Wound Matrix (SISWM) has been shown to improve healing of chronic ulcers over standard of care. In this study, we tested the hypothesis that chronic venous ulcers responsive to treatment with SISWM would more closely mimic an acute wound state as opposed to unresponsive ulcers. METHODS: Serum and wound exudate were collected at baseline and then weekly for up to 12 weeks from 12 patients receiving multiple applications of the SISWM. Levels of matrix metalloproteinases (MMP-1, MMP-2, MMP-3, MMP-9, and MMP-12), pro-inflammatory cytokines (IL-1ß, TNF-α, IL-8), and transforming growth factor beta (TGF-ß1) were evaluated. A variety of Th1/Th2 cytokines were also assayed, as were systemic anti-SIS and anti-α-gal antibody titers. RESULTS: Seven of the 12 patients eventually healed their wounds. Results showed significant decreases in MMP-1, MMP-2, MMP-3, MMP-9, TNF-α and IL-8, and significant increases in TGF-ß1 in wounds responding to treatment with the SISWM versus wounds that did not respond to treatment. None of the 12 patients formed a measurable serum antibody response to the SISWM. CONCLUSIONS: These data show that SISWM does not lead to immune system recognition or sensitization to the matrix and that wounds that went on to heal following treatment were characterized by a more acute wound state. The study confirms that the wound environment is important to healing and that turning a wound toward an acute biochemical state is key to the healing process.


Asunto(s)
Úlcera de la Pierna/terapia , Metaloproteinasas de la Matriz/administración & dosificación , Adolescente , Adulto , Exudados y Transudados/inmunología , Femenino , Humanos , Úlcera de la Pierna/sangre , Masculino , Metaloproteinasas de la Matriz/inmunología , Resultado del Tratamiento , Cicatrización de Heridas , Adulto Joven
8.
Braz Oral Res ; 33(suppl 1): e066, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31576950

RESUMEN

Considering the absence of predictable and effective therapeutic interventions for the treatment of peri-implantitis, scientific evidence concerning the host response profile around dental implants could be important for providing in the future a wider preventive and/or therapeutic window for this peri-implant lesion, indicating biomarkers that provide quantifiable measure of response to peri-implant therapy. Moreover, a better knowledge of pattern of host osteo-immunoinflammatory modulation in the presence of peri-implantitis could either benefit the early diagnostic of the disease or to cooperate to prognostic information related to the status of the peri-implant breakdown. Finally, new evidences concerning the host profile of modulators of inflammation and of osseous tissue metabolism around dental implants could explain the individual susceptibility for developing peri-implant lesions, identifying individuals or sites with increased risk for peri-implantitis. The focus of this chapter was, based on a systematically searched and critically reviewed literature, summarizing the existing knowledge in the scientific research concerning the host osteo-immunoinflammatory response to the microbiological challenge related to periimplantitis.


Asunto(s)
Implantes Dentales , Periimplantitis/inmunología , Biomarcadores , Resorción Ósea/inmunología , Interacciones Microbiota-Huesped/inmunología , Humanos , Interleucinas/inmunología , Metaloproteinasas de la Matriz/inmunología , Periimplantitis/microbiología
9.
Life Sci ; 234: 116786, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31445934

RESUMEN

Extensive degeneration of articular cartilage (AC) is a primary event in the pathogenesis of osteoarthritis (OA) and other types of joint and bone inflammation. OA results in the loss of joint function, usually accompanied by severe pain, and are the most common type of arthritis, affecting more than 10% of adults. The characteristic signs of OA are progressive cartilage destruction and, eventually, complete loss of chondrocytes. A key enzyme responsible for these degenerative changes in cartilage is matrix metalloproteinase-13 (MMP-13), which is thought to be a major contributor to the degenerative process occurring during OA pathogenesis. The aim of the present review is to shed light on the general role of MMPs, with special emphasis on MMP-13, in the induction of OA and the general basis of OA treatment. The pathogenic mechanism of this highly prevalent disease is not clear, and no effective disease-modifying treatment is currently available. Any updated information about OA treatment in human patients will also benefit companion animals such as horses and dogs, which also suffer from OA. Selective inhibition of MMP-13 seems to be an attractive therapeutic strategy.


Asunto(s)
Cartílago Articular/patología , Matriz Extracelular/patología , Metaloproteinasas de la Matriz/inmunología , Osteoartritis/patología , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/inmunología , Cartílago Articular/metabolismo , Descubrimiento de Drogas , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Humanos , Metaloproteinasa 13 de la Matriz/análisis , Metaloproteinasa 13 de la Matriz/inmunología , Metaloproteinasa 13 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Metaloproteinasas de la Matriz/análisis , Metaloproteinasas de la Matriz/metabolismo , Osteoartritis/tratamiento farmacológico , Osteoartritis/inmunología , Osteoartritis/metabolismo
10.
Biochem Biophys Res Commun ; 517(1): 132-139, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31307784

RESUMEN

Osteoarthritis (OA) is generally considered to be characterized by progressive articular cartilage destruction. Increasing evidence demonstrates that CDK9, which is a member of cyclin-dependent kinase family, plays a significant role in the regulation of acute and chronic inflammatory diseases. IL-1ß, a major proinflammatory cytokine, was used to establish a model of OA in vitro after stimulating chondrocytes. We found that CDK9 was highly expressed in in vitro and in vivo models of inflammation. The role of LDC000067 (abbreviated as LDC067), a specific inhibitor of CDK9, in protecting articular cartilage from immune response has not been fully clarified. Intriguingly, in this study, we demonstrated that LDC067 prevented IL-1ß-induced production of metalloproteinases (MMPs) and inflammatory cytokines, including MMP3, MMP9, MMP13, IL-6, IL-8 and TNF-ɑ. Furthermore, we revealed that LDC067 inhibited IL-1ß-induced NF-κB signaling pathway activation in chondrocytes. The inhibition of CDK9 could also delay cartilage degeneration in an anterior cruciate ligament transection (ACLT) mouse model in vivo. Taken together, these results highlighted the significance of this CDK9 inhibitor in preventing cartilage destruction and indicated that LDC067 might serve as a potential therapeutic agent for OA.


Asunto(s)
Condrocitos/inmunología , Quinasa 9 Dependiente de la Ciclina/inmunología , Inflamación/inmunología , Osteoartritis/inmunología , Animales , Línea Celular , Condrocitos/efectos de los fármacos , Condrocitos/patología , Quinasa 9 Dependiente de la Ciclina/análisis , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Hipertrofia/tratamiento farmacológico , Hipertrofia/inmunología , Hipertrofia/patología , Inflamación/tratamiento farmacológico , Inflamación/patología , Interleucina-1beta/análisis , Interleucina-1beta/inmunología , Masculino , Metaloproteinasas de la Matriz/análisis , Metaloproteinasas de la Matriz/inmunología , Ratones Endogámicos C57BL , FN-kappa B/análisis , FN-kappa B/inmunología , Osteoartritis/tratamiento farmacológico , Osteoartritis/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/inmunología
11.
Nat Commun ; 10(1): 2297, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-31127085

RESUMEN

Candida albicans is a fungal pathobiont, able to cause epithelial cell damage and immune activation. These functions have been attributed to its secreted toxin, candidalysin, though the molecular mechanisms are poorly understood. Here, we identify epidermal growth factor receptor (EGFR) as a critical component of candidalysin-triggered immune responses. We find that both C. albicans and candidalysin activate human epithelial EGFR receptors and candidalysin-deficient fungal mutants poorly induce EGFR phosphorylation during murine oropharyngeal candidiasis. Furthermore, inhibition of EGFR impairs candidalysin-triggered MAPK signalling and release of neutrophil activating chemokines in vitro, and diminishes neutrophil recruitment, causing significant mortality in an EGFR-inhibited zebrafish swimbladder model of infection. Investigation into the mechanism of EGFR activation revealed the requirement of matrix metalloproteinases (MMPs), EGFR ligands and calcium. We thus identify a PAMP-independent mechanism of immune stimulation and highlight candidalysin and EGFR signalling components as potential targets for prophylactic and therapeutic intervention of mucosal candidiasis.


Asunto(s)
Candida albicans/inmunología , Proteínas Fúngicas/inmunología , Interacciones Huésped-Patógeno/inmunología , Sacos Aéreos/microbiología , Animales , Candida albicans/genética , Candida albicans/metabolismo , Candidiasis/inmunología , Candidiasis/microbiología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Receptores ErbB/genética , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Femenino , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/inmunología , Metaloproteinasas de la Matriz/inmunología , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Membrana Mucosa/inmunología , Membrana Mucosa/microbiología , Faringitis/inmunología , Faringitis/microbiología , Fosforilación , Pez Cebra
12.
Int Immunopharmacol ; 72: 12-20, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30954791

RESUMEN

Inflammasomes are protein complexes that produce IL-1ß in response to damage or pathogens. As such, inflammasomes are involved in several types of hepatic fibrosis. However, the mechanisms by which these complexes drive the liver's fibrogenic status remain unclear. We co-cultured differentiated macrophages (the THP-1 cell line or human monocyte-derived macrophages (MDMs)) with human hepatic fibroblasts (either the LX-2 cell line or primary human hepatic stellate cells (HSCs)). The inflammasome pathway was activated with lipopolysaccharide (LPS) and monosodium urate (MSU) crystals, and the HSCs' responses were analyzed. Our results show that co-culture of HSCs with THP-1 cells upregulated transcription of the genes coding for metalloproteinase (MMP)-3 and MMP-9. After inflammasome pathway activation, the HSCs' phenotype was the same in the presence of THP-1 cells or MDMs (i.e. upregulation of MMP-3, MMP-9, and the pro-inflammatory cytokine IL-1ß). We found that two cytokines were involved in these changes: IL-1ß regulated MMP-3 and IL-1ß mRNA expression, whereas TNF-α regulated MMP-9 mRNA expression. Experiments with primary cells revealed that a general inflammatory environment is responsible for the downregulation of pro-fibrotic markers. Our present results suggest that inflammasome pathway activation in macrophages leads to a pro-inflammatory environment for HSCs leading to MMP/TIMP imbalance and enhanced fibrolytic properties.


Asunto(s)
Células Estrelladas Hepáticas/inmunología , Inflamasomas/inmunología , Macrófagos/inmunología , Metaloproteinasas de la Matriz/inmunología , Inhibidor Tisular de Metaloproteinasa-1/inmunología , Actinas , Células Cultivadas , Humanos , Interleucina-1beta/inmunología , Factor de Necrosis Tumoral alfa/inmunología
13.
J Immunol Methods ; 469: 26-32, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30880264

RESUMEN

Tannerella forsythia is a gram-negative anaerobic bacterium that is associated with the development of destructive periodontal disease. T. forsythia secretes the metalloprotease-like enzyme karilysin. Using in vitro systems karilysin has been shown to modulate the host immune response by degradation of complement system proteins and by inactivation of the antimicrobial peptide LL-37 by proteolytic cleavage. This makes karilysin a highly interesting virulence factor to study in the framework of drug development and diagnostics. However, to date the presence of karilysin in clinical samples has not been demonstrated due to the lack of specific probes. In the present work, a high titer and stable affinity-purified avian IgY antibody against karilysin was developed. By surface plasmon resonance imaging the IgY affinity was found to be in the low nanomolar range. The antibody could be used to detect karilysin in saliva samples by immuno-blotting and was specific when tested towards human MMP-3. Furthermore, an avian IgY-based immunoassay was developed, which demonstrated low intra- and interday assay variability (CV's below 10%). Application of the immunoassay on a well-characterized set of saliva samples from adolescents with or without signs of periodontitis showed that it was possible to detect karilysin in saliva. A significant difference in karilysin concentration was found between saliva from participants with signs of periodontitis and saliva from healthy controls (p = .0024). The median of karilysin levels among periodontitis cases was 957 pg/ml (IQR, 499-2132 pg/ml) and the median for controls was 569 pg/ml (IQR, 210-1343 pg/ml). Collectively our data confirm the presence of karilysin in clinical samples. The described IgY-based immunoassay may prove useful as part of protein-based biomarker screenings in the clinic or in point-of care settings.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/fisiología , Ensayo de Inmunoadsorción Enzimática , Infecciones por Bacterias Gramnegativas/diagnóstico , Inmunoglobulinas/inmunología , Metaloproteinasas de la Matriz/inmunología , Periodontitis/diagnóstico , Saliva/microbiología , Tannerella forsythia/inmunología , Factores de Virulencia/inmunología , Adolescente , Especificidad de Anticuerpos , Proteínas Bacterianas/inmunología , Estudios de Casos y Controles , Femenino , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Masculino , Periodontitis/microbiología , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Tannerella forsythia/patogenicidad , Virulencia
14.
Int J Mol Sci ; 20(6)2019 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-30884802

RESUMEN

Rheumatoid arthritis (RA) is a chronic, inflammatory autoimmune disease of unknown etiology. It is characterized by the presence of rheumatoid factor and anticitrullinated peptide antibodies. The orchestra of the inflammatory process among various immune cells, cytokines, chemokines, proteases, matrix metalloproteinases (MMPs), and reactive oxidative stress play critical immunopathologic roles in the inflammatory cascade of the joint environment, leading to clinical impairment and RA. With the growing understanding of the immunopathogenic mechanisms, increasingly novel marked and potential biologic agents have merged for the treatment of RA in recent years. In this review, we focus on the current understanding of pathogenic mechanisms, highlight novel biologic disease-modifying antirheumatic drugs (DMRADs), targeted synthetic DMRADs, and immune-modulating agents, and identify the applicable immune-mediated therapeutic strategies of the near future. In conclusion, new therapeutic approaches are emerging through a better understanding of the immunopathophysiology of RA, which is improving disease outcomes better than ever.


Asunto(s)
Antirreumáticos/uso terapéutico , Artritis Reumatoide/terapia , Inmunomodulación , Inflamación/terapia , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/uso terapéutico , Antirreumáticos/inmunología , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Quimiocinas/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/inmunología , Factor Reumatoide/inmunología
16.
Braz. oral res. (Online) ; 33(supl.1): e066, 2019.
Artículo en Inglés | LILACS | ID: biblio-1039322

RESUMEN

Abstract Considering the absence of predictable and effective therapeutic interventions for the treatment of peri-implantitis, scientific evidence concerning the host response profile around dental implants could be important for providing in the future a wider preventive and/or therapeutic window for this peri-implant lesion, indicating biomarkers that provide quantifiable measure of response to peri-implant therapy. Moreover, a better knowledge of pattern of host osteo-immunoinflammatory modulation in the presence of peri-implantitis could either benefit the early diagnostic of the disease or to cooperate to prognostic information related to the status of the peri-implant breakdown. Finally, new evidences concerning the host profile of modulators of inflammation and of osseous tissue metabolism around dental implants could explain the individual susceptibility for developing peri-implant lesions, identifying individuals or sites with increased risk for peri-implantitis. The focus of this chapter was, based on a systematically searched and critically reviewed literature, summarizing the existing knowledge in the scientific research concerning the host osteo-immunoinflammatory response to the microbiological challenge related to periimplantitis.


Asunto(s)
Humanos , Implantes Dentales , Periimplantitis/inmunología , Resorción Ósea/inmunología , Biomarcadores , Interleucinas/inmunología , Metaloproteinasas de la Matriz/inmunología , Periimplantitis/microbiología , Interacciones Microbiota-Huesped/inmunología
17.
Cell Death Dis ; 9(12): 1166, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518918

RESUMEN

Osteoarthritis (OA), a chronic disease characterized by articular cartilage degeneration, is a leading cause of disability and pain worldwide. In OA, chondrocytes in cartilage undergo phenotypic changes and senescence, restricting cartilage regeneration and favouring disease progression. Similar to other wound-healing disorders, chondrocytes from OA patients show a chronic increase in the gap junction channel protein connexin43 (Cx43), which regulates signal transduction through the exchange of elements or recruitment/release of signalling factors. Although immature or stem-like cells are present in cartilage from OA patients, their origin and role in disease progression are unknown. In this study, we found that Cx43 acts as a positive regulator of chondrocyte-mesenchymal transition. Overactive Cx43 largely maintains the immature phenotype by increasing nuclear translocation of Twist-1 and tissue remodelling and proinflammatory agents, such as MMPs and IL-1ß, which in turn cause cellular senescence through upregulation of p53, p16INK4a and NF-κB, contributing to the senescence-associated secretory phenotype (SASP). Downregulation of either Cx43 by CRISPR/Cas9 or Cx43-mediated gap junctional intercellular communication (GJIC) by carbenoxolone treatment triggered rediferentiation of osteoarthritic chondrocytes into a more differentiated state, associated with decreased synthesis of MMPs and proinflammatory factors, and reduced senescence. We have identified causal Cx43-sensitive circuit in chondrocytes that regulates dedifferentiation, redifferentiation and senescence. We propose that chondrocytes undergo chondrocyte-mesenchymal transition where increased Cx43-mediated GJIC during OA facilitates Twist-1 nuclear translocation as a novel mechanism involved in OA progression. These findings support the use of Cx43 as an appropriate therapeutic target to halt OA progression and to promote cartilage regeneration.


Asunto(s)
Cartílago Articular/inmunología , Comunicación Celular/genética , Senescencia Celular/genética , Condrocitos/inmunología , Conexina 43/genética , Osteoartritis/genética , Adipocitos/efectos de los fármacos , Adipocitos/inmunología , Adipocitos/patología , Antígenos CD/genética , Antígenos CD/inmunología , Carbenoxolona/farmacología , Cartílago Articular/patología , Estudios de Casos y Controles , Comunicación Celular/inmunología , Diferenciación Celular , Senescencia Celular/inmunología , Condrocitos/efectos de los fármacos , Condrocitos/patología , Conexina 43/inmunología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/inmunología , Regulación de la Expresión Génica , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/inmunología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/patología , FN-kappa B/genética , FN-kappa B/inmunología , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Osteoartritis/inmunología , Osteoartritis/patología , Cultivo Primario de Células , Índice de Severidad de la Enfermedad , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/inmunología , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/inmunología
18.
Cell Physiol Biochem ; 47(1): 11-25, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29763932

RESUMEN

BACKGROUND/AIMS: Chronic inflammation contributes to cartilage degeneration during the progression of osteoarthritis (OA). Adipose tissue-derived mesenchymal stem cells (AD-MSC) show great potential to treat inflammatory and degradative processes in OA and have demonstrated paracrine effects in chondrocytes. In the present work, we have isolated and characterized the extracellular vesicles from human AD-MSC to investigate their role in the chondroprotective actions of these cells. METHODS: AD-MSC were isolated by collagenase treatment from adipose tissue from healthy individuals subjected to abdominal lipectomy surgery. Microvesicles and exosomes were obtained from conditioned medium by filtration and differential centrifugation. Chondrocytes from OA patients were used in primary culture and stimulated with 10 ng/ml interleukin(IL)-1ß in the presence or absence of AD-MSC microvesicles, exosomes or conditioned medium. Protein expression was investigated by ELISA and immunofluorescence, transcription factor-DNA binding by ELISA, gene expression by real-time PCR, prostaglandin E2 (PGE2) by radioimmunoassay, and matrix metalloproteinase (MMP) activity and nitric oxide (NO) production by fluorometry. RESULTS: In OA chondrocytes stimulated with IL-1ß, microvesicles and exosomes reduced the production of inflammatory mediators tumor necrosis factor-α, IL-6, PGE2 and NO. The downregulation of cyclooxygenase-2 and microsomal prostaglandin E synthase-1 would lead to the decreased PGE2 production while the effect on NO could depend on the reduction of inducible nitric oxide synthase expression. Treatment of OA chondrocytes with extracellular vesicles also decreased the release of MMP activity and MMP-13 expression whereas the production of the anti-inflammatory cytokine IL-10 and the expression of collagen II were significantly enhanced. The reduction of inflammatory and catabolic mediators could be the consequence of a lower activation of nuclear factor-κB and activator protein-1. The upregulation of annexin A1 specially in MV may contribute to the anti-inflammatory and chondroprotective effects of AD-MSC. CONCLUSIONS: Our data support the interest of AD-MSC extracellular vesicles to develop new therapeutic approaches in joint conditions.


Asunto(s)
Condrocitos/inmunología , Vesículas Extracelulares/inmunología , Células Madre Mesenquimatosas/inmunología , Osteoartritis/terapia , Tejido Adiposo/citología , Anciano , Supervivencia Celular , Células Cultivadas , Condrocitos/citología , Condrocitos/patología , Citocinas/inmunología , Dinoprostona/inmunología , Femenino , Humanos , Masculino , Metaloproteinasas de la Matriz/inmunología , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Óxido Nítrico/inmunología , Osteoartritis/inmunología , Osteoartritis/patología
19.
Immunol Res ; 66(1): 59-66, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29178041

RESUMEN

Tuberculosis (TB) is a crucial public health problem with prevalence of multidrug resistant (MDR) rising. An accurate TB biomarker is urgently needed to monitor the response to treatment in patients with MDR tuberculosis. To analyze interaction between selected MDR-TB purified protein and immune cells, dendritic cells from MDR-TB patients and healthy subjects were stimulated by 55KDa protein fractions (Rv0147). The purified proteins identified by proteomic techniques (two-dimensional gel electrophoresis, mass spectrometry) and peptide sequences are known to bind a MHC class I alleles which are extracted from the Immune Epitope Database and Analysis Resource database ( www.iedb.org ). T cells were isolated from PBMC by negative selection and cells were cultured in RPMI-1640 at 37 °C and 5% CO2. Cell culture was assayed for cytokine IL-10 and INF-γ by ELISA. We found that INF-γ production was significantly (335 ± 35.5 pg/ml, P ˂ 0.05) upregulated after protein candidate (Rv0147) stimulation by dendritic cells from MDR-TB patients, whereas IL-10 production was greatly reduced compared with production in healthy subjects (212 ± 9.94 pg/ml, P ˂ 0.05). In fact, the purified protein, Rv0147, stimulated dendritic cells from MDR-TB patients, failed to produce IL-10 and directly stimulates INF-γ production by T cells. These results suggest that the purified protein, Rv0147, may stimulate Th1 type protective cytokine response in MDR-TB patients but not in normal subjects. The production of INF-γ but not IL-10 in the presence of purified protein, Rv0147, may be shifted to Th1 responses in MDR-TB patients and supports its potential as protein vaccine candidates against TB.


Asunto(s)
Proteínas Bacterianas/inmunología , Células Dendríticas/inmunología , Epítopos Inmunodominantes/inmunología , Metaloproteinasas de la Matriz/inmunología , Mycobacterium tuberculosis/fisiología , Células TH1/inmunología , Tuberculosis/inmunología , Biomarcadores , Células Cultivadas , Técnicas de Cocultivo , Resistencia a Múltiples Medicamentos , Humanos , Inmunidad Celular , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Activación de Linfocitos , Vacunas contra la Tuberculosis/inmunología
20.
J Eur Acad Dermatol Venereol ; 31 Suppl 5: 8-12, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28805938

RESUMEN

Acne is a chronic inflammatory disease of the pilosebaceous unit. Its pathophysiology includes hyperseborrhoea, abnormal follicular keratinization and Propionibacterium acnes proliferation in the pilosebaceous unit. Recent research has shed some new light on the involvement of the sebaceous gland, as well as on the pro-inflammatory activity of the cutaneous microbiome. During puberty, alteration of the sebaceous lipid profile, called dysseborrhoea, stress, irritation, cosmetics and potential dietary factors lead to inflammation and formation of different types of acne lesions. Dysbiosis, the process leading to a disturbed skin barrier and disequilibrium of the cutaneous microbiome, resulting in the proliferation of P. acnes strains, is another important process that triggers acne. P. acnes activates the innate immunity via the expression of protease activated receptors (PARs), tumour necrosis factor (TNF) α and toll-like receptors (TLRs), and the production of interferon (INF) γ, interleukins (IL-8, IL12, IL-1), TNF, and matrix metalloproteinases (MMPs) by keratinocytes, resulting in the hyperkeratinization of the pilosebaceous unit. Rebalancing the natural microbiome of the skin by restoring the natural skin barrier, limiting the proliferation of P. acnes on the skin by using topical antibacterials which do not cause resistance and regulating quantity and quality of sebum will be the main acne treatment challenges in the future. The aim of this article to provide an update on the involvement of the sebaceous gland, the innate immunity and the cutaneous microbiome, how all of these factors promote acne and to illustrate their links with current and future treatments.


Asunto(s)
Acné Vulgar/patología , Acné Vulgar/inmunología , Acné Vulgar/microbiología , Biopelículas , Citocinas/inmunología , Endocannabinoides/fisiología , Humanos , Inmunidad Innata , Metaloproteinasas de la Matriz/inmunología , Propionibacterium acnes/patogenicidad , Receptores de Superficie Celular/inmunología , Glándulas Sebáceas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA