Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Hum Pathol ; 135: 22-34, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36871865

RESUMEN

Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) is a rare autosomal dominant disease resulting from a frame-shift mutation in TREX1, an intracellular 3'-5' exonuclease 1. Hepatic findings include an elevated alkaline phosphatase (ALP) and nodular regenerative hyperplasia (NRH). Affected individuals typically succumb to brain lesions before clinically apparent hepatic manifestations; thus, little else is known about the hepatic pathology. Autopsy reports and a liver section from each (n = 11) of three unrelated kindreds with the most common mutation in TREX1 (V235Gfs∗6) were studied with standard and immunohistochemical stains. Cases were compared with "normal liver" controls from similar autopsy years. Cases consisted of six men and five women who died at a median age of 50 yr (range, 41-60 yr.). Seven had elevated ALP. Two had liver atrophy. Foci of NRH were variably detected in all. Inhomogeneous distribution of other findings included patternless parenchymal fibrous bands, approximation of vascular structures, and commonly, architectural changes of vascular structures. Only bile duct epithelia were unaffected. In addition, small trichrome-positive nodules were found along vein walls or isolated in the parenchyma. Rare foci of non-NRH hepatocytic nodules were noted in 3. Increased CD34 and altered α-SMA IHC expression were variably noted. Periportal ductules and perivenular K7 IHC expression were increased to unpredictable degrees. The extensive but inhomogeneous histopathologic findings in livers of autopsied patients with RVCL-S appear to involve hepatic vascular structures. These findings validate inclusion of vascular liver involvement beyond NRH in this complex hereditary disorder.


Asunto(s)
Leucoencefalopatías , Hepatopatías , Enfermedades Vasculares , Masculino , Humanos , Femenino , Hiperplasia/patología , Hígado/patología , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología , Hepatopatías/genética , Hepatopatías/patología
2.
Immunol Rev ; 313(1): 60-63, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36089772

RESUMEN

Primitive underpinnings of the alternative pathway (AP), namely, a C3-like protein, likely arose more than a billion years ago. The development of an AP amplification loop, while greatly enhancing speed and potency, also presents a double-edged sword. Although critical to combat an infectious disease, it is also potentially destructive, particularly in a chronic disease process involving vital organs where scarring and reduction of regulatory function can occur. Furthermore, new knowledge is pointing to genetic factors involved in an increasing number of complement-related diseases such as age-related macular degeneration. However, even a normal functioning repertoire of complement components can drive cellular damage as a result of low-level complement activation over time. Thus, the modern human AP now faces a new challenge: cumulatively-driven tissue damage from chronic inflammatory processes that mediate cellular injury. The impact of ongoing low-level AP-enhanced complement activation in disease processes is just beginning to be appreciated and studied. However, the sheer numbers of individuals affected by chronic diseases emphasize the need for novel therapeutic agents capable of modulating the AP. The more we learn about this ancient system, the greater is the likelihood of developing fresh perspectives that could contribute to improved human health.


Asunto(s)
Activación de Complemento , Vía Alternativa del Complemento , Humanos , Vía Alternativa del Complemento/genética , Proteínas del Sistema Complemento/metabolismo , Inflamación
3.
J Virol ; 96(3): e0082621, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-34787457

RESUMEN

Human adenovirus serotype 26 (Ad26) is used as a gene-based vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and HIV-1. However, its primary receptor portfolio remains controversial, potentially including sialic acid, coxsackie and adenovirus receptor (CAR), integrins, and CD46. We and others have shown that Ad26 can use CD46, but these observations were questioned on the basis of the inability to cocrystallize Ad26 fiber with CD46. Recent work demonstrated that Ad26 binds CD46 with its hexon protein rather than its fiber. We examined the functional consequences of Ad26 for infection in vitro and in vivo. Ectopic expression of human CD46 on Chinese hamster ovary cells increased Ad26 infection significantly. Deletion of the complement control protein domain CCP1 or CCP2 or the serine-threonine-proline (STP) region of CD46 reduced infection. Comparing wild-type and sialic acid-deficient CHO cells, we show that the usage of CD46 is independent of its sialylation status. Ad26 transduction was increased in CD46 transgenic mice after intramuscular (i.m.) injection but not after intranasal (i.n.) administration. Ad26 transduction was 10-fold lower than Ad5 transduction after intratumoral (i.t.) injection of CD46-expressing tumors. Ad26 transduction of liver was 1,000-fold lower than that ofAd5 after intravenous (i.v.) injection. These data demonstrate the use of CD46 by Ad26 in certain situations but also show that the receptor has little consequence by other routes of administration. Finally, i.v. injection of high doses of Ad26 into CD46 mice induced release of liver enzymes into the bloodstream and reduced white blood cell counts but did not induce thrombocytopenia. This suggests that Ad26 virions do not induce direct clotting side effects seen during coronavirus disease 2019 (COVID-19) vaccination with this serotype of adenovirus. IMPORTANCE The human species D Ad26 is being investigated as a low-seroprevalence vector for oncolytic virotherapy and gene-based vaccination against HIV-1 and SARS-CoV-2. However, there is debate in the literature about its tropism and receptor utilization, which directly influence its efficiency for certain applications. This work was aimed at determining which receptor(s) this virus uses for infection and its role in virus biology, vaccine efficacy, and, importantly, vaccine safety.


Asunto(s)
Infecciones por Adenovirus Humanos/metabolismo , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/clasificación , Adenovirus Humanos/fisiología , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Interacciones Huésped-Patógeno , Proteína Cofactora de Membrana/metabolismo , Adenovirus Humanos/ultraestructura , Animales , Biomarcadores , Recuento de Células Sanguíneas , Células CHO , Línea Celular , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/química , Cricetulus , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Proteína Cofactora de Membrana/química , Proteína Cofactora de Membrana/genética , Ratones Transgénicos , Modelos Biológicos , Modelos Moleculares , Mutagénesis , Unión Proteica , Conformación Proteica , Serogrupo , Ácidos Siálicos/metabolismo , Ácidos Siálicos/farmacología , Relación Estructura-Actividad
4.
Viruses ; 13(7)2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34202570

RESUMEN

Dengue is a mosquito-borne viral disease causing significant health and economic burdens globally. The dengue virus (DENV) comprises four serotypes (DENV1-4). Usually, the primary infection is asymptomatic or causes mild dengue fever (DF), while secondary infections with a different serotype increase the risk of severe dengue disease (dengue hemorrhagic fever, DHF). Complement system activation induces inflammation and tissue injury, contributing to disease pathogenesis. However, in asymptomatic or primary infections, protective immunity largely results from the complement system's lectin pathway (LP), which is activated through foreign glycan recognition. Differences in N-glycans displayed on the DENV envelope membrane influence the lectin pattern recognition receptor (PRR) binding efficiency. The important PRR, mannan binding lectin (MBL), mediates DENV neutralization through (1) a complement activation-independent mechanism via direct MBL glycan recognition, thereby inhibiting DENV attachment to host target cells, or (2) a complement activation-dependent mechanism following the attachment of complement opsonins C3b and C4b to virion surfaces. The serum concentrations of lectin PRRs and their polymorphisms influence these LP activities. Conversely, to escape the LP attack and enhance the infectivity, DENV utilizes the secreted form of nonstructural protein 1 (sNS1) to counteract the MBL effects, thereby increasing viral survival and dissemination.


Asunto(s)
Lectina de Unión a Manosa de la Vía del Complemento , Virus del Dengue/inmunología , Virus del Dengue/patogenicidad , Dengue/inmunología , Dengue/virología , Animales , Humanos , Evasión Inmune , Lectina de Unión a Manosa/sangre , Lectina de Unión a Manosa/genética , Lectina de Unión a Manosa/inmunología , Lectina de Unión a Manosa/metabolismo , Polimorfismo de Nucleótido Simple , Polisacáridos/inmunología , Polisacáridos/metabolismo , Receptores de Reconocimiento de Patrones/sangre , Receptores de Reconocimiento de Patrones/genética , Receptores de Reconocimiento de Patrones/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Dengue Grave/inmunología , Dengue Grave/virología , Proteínas no Estructurales Virales/metabolismo , Virulencia
5.
Mult Scler Relat Disord ; 52: 103015, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34044261

RESUMEN

Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) is a rare fatal autosomal dominant vasculopathy associated with mutations in the TREX1 gene. Only one de novo case has been reported in the literature. We report the long-term clinical, radiological, and pathological presentation of a patient with a de novo and novel mutation in this gene. Description of the clinical, genetic, imaging and pathologic characteristics is important to better characterize RVCL-S and prevent unnecessary interventions. RVCL-S should be considered in patients with tumefactive brain lesions unresponsive to immunotherapy.


Asunto(s)
Enfermedades Desmielinizantes , Leucoencefalopatías , Enfermedades Vasculares , Exodesoxirribonucleasas , Humanos , Mutación , Fosfoproteínas
6.
Curr Opin Immunol ; 72: 126-134, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34004375

RESUMEN

Membrane cofactor protein (MCP; CD46), a ubiquitously expressed complement regulatory protein, serves as a cofactor for serine protease factor I to cleave and inactivate C3b and C4b deposited on host cells. However, CD46 also plays roles in human reproduction, autophagy, modulating T cell activation and effector functions and is a member of the newly identified intracellular complement system (complosome). CD46 also is a receptor for 11 pathogens ('pathogen magnet'). While CD46 deficiencies contribute to inflammatory disorders, its overexpression in cancers and role as a receptor for some adenoviruses has led to its targeting by oncolytic agents and adenoviral-based therapeutic vectors, including coronavirus disease of 2019 (COVID-19) vaccines. This review focuses on recent advances in identifying disease-causing CD46 variants and its pathogen connections.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Proteína Cofactora de Membrana/metabolismo , SARS-CoV-2/fisiología , Linfocitos T/inmunología , Animales , Autofagia , Activación de Complemento , Interacciones Huésped-Patógeno , Humanos , Activación de Linfocitos , Proteína Cofactora de Membrana/genética , Viroterapia Oncolítica , Polimorfismo Genético , Reproducción
7.
Antibodies (Basel) ; 9(4)2020 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-33147799

RESUMEN

One of the most challenging aspects of cancer therapeutics is target selection. Recently, CD46 (membrane cofactor protein; MCP) has emerged as a key player in both malignant transformation as well as in cancer treatments. Normally a regulator of complement activation, CD46 is co-expressed as four predominant isoforms on almost all cell types. CD46 is highly overexpressed on a variety of human tumor cells. Clinical and experimental data support an association between increased CD46 expression and malignant transformation and metastasizing potential. Further, CD46 is a newly discovered driver of metabolic processes and plays a role in the intracellular complement system (complosome). CD46 is also known as a pathogen magnet due to its role as a receptor for numerous microbes, including several species of measles virus and adenoviruses. Strains of these two viruses have been exploited as vectors for the therapeutic development of oncolytic agents targeting CD46. In addition, monoclonal antibody-drug conjugates against CD46 also are being clinically evaluated. As a result, there are multiple early-phase clinical trials targeting CD46 to treat a variety of cancers. Here, we review CD46 relative to these oncologic connections.

8.
Neurology ; 95(14): e1918-e1931, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32887784

RESUMEN

OBJECTIVE: To characterize lesion evolution and neurodegeneration in retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) using multimodal MRI. METHODS: We prospectively performed MRI and cognitive testing in RVCL-S and healthy control cohorts. Gray and white matter volume and disruption of white matter microstructure were quantified. Asymmetric spin echo acquisition permitted voxel-wise oxygen extraction fraction (OEF) calculation as an in vivo marker of microvascular ischemia. The RVCL-S cohort was included in a longitudinal analysis of lesion subtypes in which hyperintense lesions on fluid-attenuated inversion recovery (FLAIR), T1-postgadolinium, and diffusion-weighted imaging were delineated and quantified volumetrically. RESULTS: Twenty individuals with RVCL-S and 26 controls were enrolled. White matter volume and microstructure declined faster in those with RVCL-S compared to controls. White matter atrophy in RVCL-S was highly linear (ρ = -0.908, p < 0.0001). Normalized OEF was elevated in RVCL-S and increased with disease duration. Multiple cognitive domains, specifically those measuring working memory and processing speed, were impaired in RVCL-S. Lesion volumes, regardless of subtype, progressed/regressed with high variability as a function of age, while FLAIR lesion burden increased near time to death (p < 0.001). CONCLUSION: RVCL-S is a monogenic microvasculopathy affecting predominantly the white matter with regard to atrophy and cognitive impairment. White matter volumes in RVCL-S declined linearly, providing a potential metric against which to test the efficacy of future therapies. Progressive elevation of white matter OEF suggests that microvascular ischemia may underlie neurodegeneration in RVCL-S.


Asunto(s)
Disfunción Cognitiva/patología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Degeneración Nerviosa/patología , Enfermedades de la Retina/patología , Enfermedades Vasculares/patología , Sustancia Blanca/patología , Adulto , Disfunción Cognitiva/diagnóstico por imagen , Femenino , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Degeneración Nerviosa/diagnóstico por imagen , Neuroimagen/métodos , Enfermedades de la Retina/diagnóstico por imagen , Enfermedades Vasculares/diagnóstico por imagen , Sustancia Blanca/diagnóstico por imagen
9.
JCI Insight ; 5(15)2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32554923

RESUMEN

Coronavirus disease 2019 (COVID-19), the disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has resulted in a global pandemic and a disruptive health crisis. COVID-19-related morbidity and mortality have been attributed to an exaggerated immune response. The role of complement activation and its contribution to illness severity is being increasingly recognized. Here, we summarize current knowledge about the interaction of coronaviruses with the complement system. We posit that (a) coronaviruses activate multiple complement pathways; (b) severe COVID-19 clinical features often resemble complementopathies; (c) the combined effects of complement activation, dysregulated neutrophilia, endothelial injury, and hypercoagulability appear to be intertwined to drive the severe features of COVID-19; (d) a subset of patients with COVID-19 may have a genetic predisposition associated with complement dysregulation; and (e) these observations create a basis for clinical trials of complement inhibitors in life-threatening illness.


Asunto(s)
Betacoronavirus , Activación de Complemento/inmunología , Infecciones por Coronavirus , Pandemias , Neumonía Viral , Betacoronavirus/patogenicidad , Betacoronavirus/fisiología , Coagulación Sanguínea , COVID-19 , Infecciones por Coronavirus/sangre , Infecciones por Coronavirus/inmunología , Células Endoteliales/virología , Humanos , Neumonía Viral/sangre , Neumonía Viral/inmunología , SARS-CoV-2
12.
Front Immunol ; 10: 703, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31019515

RESUMEN

Discovery of a C3(H2O) uptake pathway has led to renewed interest in this alternative pathway triggering form of C3 in human biospecimens. Previously, a quantifiable method to measure C3(H2O), not confounded by other complement activation products, was unavailable. Herein, we describe a sensitive and specific ELISA for C3(H2O). We initially utilized this assay to determine baseline C3(H2O) levels in healthy human fluids and to define optimal sample storage and handling conditions. We detected ~500 ng/ml of C3(H2O) in fresh serum and plasma, a value substantially lower than what was predicted based on previous studies with purified C3 preparations. After a single freeze-thaw cycle, the C3(H2O) concentration increased 3- to 4-fold (~2,000 ng/ml). Subsequent freeze-thaw cycles had a lesser impact on C3(H2O) generation. Further, we found that storage of human sera or plasma samples at 4°C for up to 22 h did not generate additional C3(H2O). To determine the potential use of C3(H2O) as a biomarker, we evaluated specimens from patients with inflammatory-driven diseases. C3(H2O) concentrations were moderately increased (1.5- to 2-fold) at baseline in sera from active systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) patients compared to healthy controls. In addition, upon challenge with multiple freeze-thaw cycles or incubation at 22 or 37°C, C3(H2O) generation was significantly enhanced in SLE and RA patients' sera. In bronchoalveolar lavage fluid from lung-transplant recipients, we noted a substantial increase in C3(H2O) within 3 months of acute antibody-mediated rejection. In conclusion, we have established an ELISA for assessing C3(H2O) as a diagnostic and prognostic biomarker in human diseases.


Asunto(s)
Complemento C3/análisis , Ensayo de Inmunoadsorción Enzimática/métodos , Inflamación/sangre , Inflamación/inmunología , Artritis Reumatoide/sangre , Artritis Reumatoide/inmunología , Biomarcadores/sangre , Líquido del Lavado Bronquioalveolar/inmunología , Estudios de Casos y Controles , Activación de Complemento , Rechazo de Injerto/sangre , Rechazo de Injerto/inmunología , Humanos , Trasplante de Pulmón/efectos adversos , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Pronóstico , Agua/análisis
13.
J Clin Invest ; 129(3): 1061-1075, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30714990

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is frequently associated in humans with loss-of-function mutations in complement-regulating proteins or gain-of-function mutations in complement-activating proteins. Thus, aHUS provides an archetypal complement-mediated disease with which to model new therapeutic strategies and treatments. Herein, we show that, when transferred to mice, an aHUS-associated gain-of-function change (D1115N) to the complement-activation protein C3 results in aHUS. Homozygous C3 p.D1115N (C3KI) mice developed spontaneous chronic thrombotic microangiopathy together with hematuria, thrombocytopenia, elevated creatinine, and evidence of hemolysis. Mice with active disease had reduced plasma C3 with C3 fragment and C9 deposition within the kidney. Therapeutic blockade or genetic deletion of C5, a protein downstream of C3 in the complement cascade, protected homozygous C3KI mice from thrombotic microangiopathy and aHUS. Thus, our data provide in vivo modeling evidence that gain-of-function changes in complement C3 drive aHUS. They also show that long-term C5 deficiency is not accompanied by development of other renal complications (such as C3 glomerulopathy) despite sustained dysregulation of C3. Our results suggest that this preclinical model will allow testing of novel complement inhibitors with the aim of developing precisely targeted therapeutics that could have application in many complement-mediated diseases.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Activación de Complemento , Complemento C3 , Complemento C5 , Riñón , Mutación Missense , Sustitución de Aminoácidos , Animales , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/inmunología , Síndrome Hemolítico Urémico Atípico/patología , Complemento C3/genética , Complemento C3/inmunología , Complemento C5/genética , Complemento C5/inmunología , Complemento C9/genética , Complemento C9/inmunología , Modelos Animales de Enfermedad , Glomerulonefritis Membranosa/genética , Glomerulonefritis Membranosa/inmunología , Glomerulonefritis Membranosa/patología , Riñón/inmunología , Riñón/patología , Ratones , Ratones Transgénicos
14.
J Biol Chem ; 294(13): 4878-4888, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30670593

RESUMEN

ER protein 57 (ERp57), a thiol isomerase secreted from vascular cells, is essential for complete thrombus formation in vivo, but other extracellular ERp57 functions remain unexplored. Here, we employed a kinetic substrate-trapping approach to identify extracellular protein substrates of ERp57 in platelet-rich plasma. MS-based identification with immunochemical confirmation combined with gene ontology enrichment analysis revealed that ERp57 targets, among other substrates, components of the lectin pathway of complement activation: mannose-binding lectin, ficolin-2, ficolin-3, collectin-10, collectin-11, mannose-binding lectin-associated serine protease-1, and mannose-binding lectin-associated serine protease-2. Ficolin-3, the most abundant lectin pathway initiator in humans, circulates as disulfide-linked multimers of a monomer. ERp57 attenuated ficolin-3 ligand recognition and complement activation by cleaving intermolecular disulfide bonds in large ficolin-3 multimers, thereby reducing multimer size and ligand-binding affinity. We used MS to identify the disulfide-bonding pattern in ficolin-3 multimers and the disulfide bonds targeted by ERp57 and found that Cys6 and Cys23 in the N-terminal region of ficolin-3 form the intermolecular disulfide bonds in ficolin-3 multimers that are reduced by ERp57. Our results not only demonstrate that ERp57 can negatively regulate complement activation, but also identify a control mechanism for lectin pathway initiation in the vasculature. We conclude that extensive multimerization in large ficolin-3 multimers leads to a high affinity for ligands and strong complement-activating potential and that ERp57 suppresses complement activation by cleaving disulfide bonds in ficolin-3 and reducing its multimer size.


Asunto(s)
Lectina de Unión a Manosa de la Vía del Complemento , Glicoproteínas/metabolismo , Lectinas/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Multimerización de Proteína , Proteolisis , Glicoproteínas/genética , Humanos , Lectinas/genética , Proteína Disulfuro Isomerasas/genética
15.
Am J Respir Cell Mol Biol ; 60(2): 144-157, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30156437

RESUMEN

The complement system provides host defense against pathogens and environmental stress. C3, the central component of complement, is present in the blood and increases in BAL fluid after injury. We recently discovered that C3 is taken up by certain cell types and cleaved intracellularly to C3a and C3b. C3a is required for CD4+ T-cell survival. These observations made us question whether complement operates at environmental interfaces, particularly in the respiratory tract. We found that airway epithelial cells (AECs, represented by both primary human tracheobronchial cells and BEAS-2B [cell line]) cultured in C3-free media were unique from other cell types in that they contained large intracellular stores of de novo synthesized C3. A fraction of this protein reduced ("storage form") but the remainder did not, consistent with it being pro-C3 ("precursor form"). These two forms of intracellular C3 were absent in CRISPR knockout-induced C3-deficient AECs and decreased with the use of C3 siRNA, indicating endogenous generation. Proinflammatory cytokine exposure increased both stored and secreted forms of C3. Furthermore, AECs took up C3 from exogenous sources, which mitigated stress-associated cell death (e.g., from oxidative stress or starvation). C3 stores were notably increased within AECs in lung tissues from individuals with different end-stage lung diseases. Thus, at-risk cells furnish C3 through biosynthesis and/or uptake to increase locally available C3 during inflammation, while intracellularly, these stores protect against certain inducers of cell death. These results establish the relevance of intracellular C3 to airway epithelial biology and suggest novel pathways for complement-mediated host protection in the airway.


Asunto(s)
Bronquios/citología , Complemento C3/metabolismo , Células Epiteliales/fisiología , Muerte Celular , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Complemento C3/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Caliciformes/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Estrés Fisiológico
17.
J Clin Invest ; 127(3): 970-981, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28192370

RESUMEN

An intracellular complement system (ICS) has recently been described in immune and nonimmune human cells. This system can be activated in a convertase-independent manner from intracellular stores of the complement component C3. The source of these stores has not been rigorously investigated. In the present study, Western blotting identified a band corresponding to C3 in freshly isolated human peripheral blood cells that was absent in corresponding cell lines. One difference between native cells and cell lines was the time absent from a fluid-phase complement source; therefore, we hypothesized that loading C3 from plasma was a route of establishing intracellular C3 stores. We found that many types of human cells specifically internalized C3(H2O), the hydrolytic product of C3, and not native C3, from the extracellular milieu. Uptake was rapid, saturable, and sensitive to competition with unlabeled C3(H2O), indicating a specific mechanism of loading. Under steady-state conditions, approximately 80% of incorporated C3(H2O) was returned to the extracellular space. These studies identify an ICS recycling pathway for C3(H2O). The loaded C3(H2O) represents a source of C3a, and its uptake altered the cytokine profile of activated CD4+ T cells. Importantly, these results indicate that the impact of soluble plasma factors should be considered when performing in vitro studies assessing cellular immune function.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Complemento C3/metabolismo , Activación de Linfocitos , Proteolisis , Linfocitos T CD4-Positivos/citología , Línea Celular , Femenino , Humanos , Masculino
18.
Mol Immunol ; 84: 2-9, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28196665

RESUMEN

A key component of both innate and adaptive immunity, new understandings of the complement system are expanding its roles beyond that traditionally appreciated. Evidence is accumulating that complement has an intracellular arsenal of components that provide not only immune defense, but also assist in key interactions for host cell functions. Although early work has primarily centered on T cells, the intracellular complement system likely functions in many if not most cells of the body. Some of these functions may trace their origins to the primitive complement system that began as a primeval form of C3 likely tasked for protection from intracellular pathogen invasion. This later expanded to include extracellular defense as C3 became a secreted protein to patrol the vasculature. Other components were added to the growing system including regulators to protect host cells from the indiscriminate effects of this potent system. Contemporary cells may retain some of these vestigial remnants. We now know that a) C3 serves as a damage-associated molecular pattern (in particular by coating pathogens that translocate into cells), b) most cells store C3 and recycle C3(H2O) for immediate use, and c) C3 assists in cellular survival and metabolic reprogramming. Other components also are part of this hidden arsenal including C5, properdin, factors H and B, and complement receptors. Importantly, better definition of the intracellular complement system may translate into new target discovery to assist in creating the next generation of complement therapeutics.


Asunto(s)
Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Animales , Humanos
19.
Annu Rev Pathol ; 12: 25-52, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-27959629

RESUMEN

The vertebrate complement system consists of sequentially interacting proteins that provide for a rapid and powerful host defense. Nearly 60 proteins comprise three activation pathways (classical, alternative, and lectin) and a terminal cytolytic pathway common to all. Attesting to its potency, nearly half of the system's components are engaged in its regulation. An emerging theme over the past decade is that variations in these inhibitors predispose to two scourges of modern humans. One, occurring most often in childhood, is a rare but deadly thrombomicroangiopathy called atypical hemolytic uremic syndrome. The other, age-related macular degeneration, is the most common form of blindness in the elderly. Their seemingly unrelated clinical presentations and pathologies share the common theme of overactivity of the complement system's alternative pathway. This review summarizes insights gained from contemporary genetics for understanding how dysregulation of this powerful innate immune system leads to these human diseases.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/fisiopatología , Vía Alternativa del Complemento/genética , Proteínas del Sistema Complemento/genética , Degeneración Macular/fisiopatología , Genotipo , Humanos
20.
Immunol Rev ; 274(1): 9-15, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27782327

RESUMEN

The complement system is an evolutionarily ancient component of immunity that revolves around the central component C3. With the recent description of intracellular C3 stores in many types of human cells, our view of the complement system has expanded. In this article, we hypothesize that a primitive version of C3 comprised the first element of the original complement system and initially functioned intracellularly and on the membrane of single-celled organisms. With increasing specialization and multicellularity, C3 evolved a secretory capacity that allowed it to play a protective role in the interstitial space. Upon development of a pumped circulatory system, C3 was synthesized in large amounts and secreted by the liver to protect the intravascular space. Recent discoveries of intracellular C3 activation, a C3-based recycling pathway and C3 being a driver and programmer of cell metabolism suggest that the complement system utilizes C3 to guard not only extracellular but also the intracellular environment. We predict that the major functions of C3 in all four locations (i.e. intracellular, membrane, interstitium and circulation) are similar: opsonization, membrane perturbation, triggering inflammation, and metabolic reprogramming.


Asunto(s)
Evolución Biológica , Vasos Sanguíneos/fisiología , Complemento C3/inmunología , Inflamación/inmunología , Hígado/fisiología , Animales , Membrana Celular/metabolismo , Activación de Complemento , Espacio Extracelular , Humanos , Inmunidad , Espacio Intracelular , Modelos Inmunológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...