Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(5)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38473963

RESUMEN

The protein p32 (C1QBP) is a multifunctional and multicompartmental homotrimer that is overexpressed in many cancer types, including colon cancer. High expression levels of C1QBP are negatively correlated with the survival of patients. Previously, we demonstrated that C1QBP is an essential promoter of migration, chemoresistance, clonogenic, and tumorigenic capacity in colon cancer cells. However, the mechanisms underlying these functions and the effects of specific C1QBP protein inhibitors remain unexplored. Here, we show that the specific pharmacological inhibition of C1QBP with the small molecule M36 significantly decreased the viability rate, clonogenic capacity, and proliferation rate of different colon cancer cell lines in a dose-dependent manner. The effects of the inhibitor of C1QBP were cytostatic and non-cytotoxic, inducing a decreased activation rate of critical pro-malignant and mitogenic cellular pathways such as Akt-mTOR and MAPK in RKO colon cancer cells. Additionally, treatment with M36 significantly affected the mitochondrial integrity and dynamics of malignant cells, indicating that p32/C1QBP plays an essential role in maintaining mitochondrial homeostasis. Altogether, our results reinforce that C1QBP is an important oncogene target and that M36 may be a promising therapeutic drug for the treatment of colon cancer.


Asunto(s)
Neoplasias del Colon , Citostáticos , Humanos , Citostáticos/farmacología , Mitógenos/farmacología , Transducción de Señal , Proteínas Mitocondriales/metabolismo , Proliferación Celular , Proteínas Portadoras/metabolismo
2.
J Virol ; 96(19): e0095922, 2022 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-36121300

RESUMEN

PCV2 has been reported to reduce the protective effects of various vaccines on immunized pigs. Our previous studies showed that the interaction of Cap and host protein gC1qR mediated the PCV2 infection-induced suppression of immune response. Thus, we wondered whether the gC1qR binding site mutant PCV2RmA could be a vaccine strain and whether this mutant PCV2RmA impairs other vaccines. Herein, we showed that PCV2 infection reduced the classic swine fever virus (CSFV) vaccine-induced generation of memory CD4+ T cells through the interaction of Cap with gC1qR. PCV2RmA can effectively induce the production of PCV2-specific antibodies, neutralizing antibodies, and peripheral blood lymphocyte proliferation in piglets at the same levels as the commercial inactivated PCV2 vaccine. The PCV2RmA-induced anti-PCV2 immune responses could eliminate the serum virus and would not lead to pathological lesions like wild-type PCV2. Moreover, compared to the commercial inactivated PCV2 vaccine, PCV2RmA is capable of inducing more durable protective immunity against PCV2 that induced production of PCV2-specific antibodies and neutralizing antibodies for a longer time via stronger induction of memory CD4+ T cells. Importantly, PCV2RmA infection did not impair the CSFV vaccine-induced generation of memory CD4+ T cells. Collectively, our findings showed that PCV2 infection impairs memory CD4+ T-cell generation to affect vaccination and provide evidence for the use of PCV2RmA as an efficient vaccine to prevent PCV2 infection. IMPORTANCE PCV2 is one of the costliest pathogens in pigs worldwide. Usage of PCV2 vaccines can prevent the PCV2 infection-induced clinical syndromes but not the viral spread. Our previous work found that PCV2 infection suppresses the host type I interferon innate immune response and CD4+ T-cell-mediated Th1 immune response through the interaction of Cap with host gC1qR. Here, we showed that the gC1qR binding site mutant PCV2RmA could effectively induce anti-PCV2 immunity and provide more durable protective immunity against wild-type PCV2 infection in pigs. PCV2RmA would not impair the generation of memory CD4+ T cells induced by classic swine fever virus (CSFV) vaccines as wild-type PCV2 did. Therefore, PCV2RmA can serve as a potential vaccine strain to better protect pigs against PCV2 infection.


Asunto(s)
Linfocitos T CD4-Positivos , Virus de la Fiebre Porcina Clásica , Peste Porcina Clásica , Receptores de Complemento , Vacunas Virales , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Sitios de Unión , Linfocitos T CD4-Positivos/inmunología , Proteínas de la Cápside/genética , Peste Porcina Clásica/inmunología , Peste Porcina Clásica/prevención & control , Peste Porcina Clásica/virología , Virus de la Fiebre Porcina Clásica/genética , Memoria Inmunológica , Interferón Tipo I , Receptores de Complemento/metabolismo , Porcinos , Vacunas de Productos Inactivados/genética , Vacunas Virales/genética
3.
Semin Immunol ; 45: 101338, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31744753

RESUMEN

In the past several years, a number of C1q binding surface proteins or receptors have been described. This is not of course surprising considering the complexity of the C1q molecule and its ability to bind to a wide range of cellular and plasma proteins via both its collagen-like [cC1q] region and its heterotrimeric globular heads [gC1q] each of which in turn is capable of binding a specific ligand. However, while each of these "receptor" molecules undoubtedly plays a specific function within its restricted microenvironment, and therefore merits full attention, this review nonetheless, will singularly focus on the structure and function of gC1qR-a multi-functional and multi-compartmental protein, which plays an important role in inflammation, infection, and cancer. Although first identified as a receptor for C1q, gC1qR has been shown to bind to a plethora of proteins found in plasma, on the cell surface and on pathogenic microorganisms. The plasma proteins that bind to gC1qR are mostly blood coagulation proteins and include high molecular weight kininogen [HK], Factor XII [Hageman factor], fibrinogen, thrombin [FII], and multimeric vitronectin. This suggests that gC1qR can play an important role in modulating not only of fibrin formation, particularly at local sites of immune injury and/or inflammation, but by activating the kinin/kallikrein system, it is also able to generate, bradykinin, a powerful vasoactive peptide that is largely responsible for the swelling seen in angioedema. Another important function of gC1qR is in cancer, where it has been shown to play a role in tumor cell survival, growth and metastatic invasion by interacting with critical molecules in the tumor cell microenvironment including those of the complement system and kinin system. Finally, by virtue of its ability to interact with a growing list of pathogen-associated molecules, including bacterial and viral ligands, gC1qR is becoming recognized as an important pathogen recognition receptor [PRR]. Given the numerous roles it plays in a growing list of disease settings, gC1qR has now become a potential target for the development of monoclonal antibody-based and/or small molecule-based therapies.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Receptores de Complemento/metabolismo , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Mapeo Cromosómico , Susceptibilidad a Enfermedades , Interacciones Huésped-Parásitos , Interacciones Huésped-Patógeno , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Estructura Molecular , Receptores de Complemento/química , Receptores de Complemento/genética , Relación Estructura-Actividad
4.
Int J Mol Sci ; 24(8)2023 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-37108418

RESUMEN

Listeria monocytogenes virulence factor InlB specifically interacts with the receptors c-Met and gC1q-R. Both receptors are present in non-professional and professional phagocytes, including macrophages. Phylogenetically defined InlB isoforms differently support invasion into non-professional phagocytes. This work deals with the effects of InlB isoforms on L. monocytogenes uptake and intracellular proliferation in human macrophages. Three isoforms of the receptor binding domain (idInlB) were derived from phylogenetically distinct L. monocytogenes strains belonging to the highly virulent CC1 (idInlBCC1), medium-virulence CC7 (idInlBCC7), and low-virulence CC9 (idInlBCC9) clonal complexes. The constant dissociation increased in the order idInlBCC1 << idInlBCC7 < idInlBCC9 for interactions with c-Met, and idInlBCC1 ≈ idInlBCC7 < idInlBCC9 for interactions with gC1q-R. The comparison of uptake and intracellular proliferation of isogenic recombinant strains which expressed full-length InlBs revealed that the strain expressing idInlBCC1 proliferated in macrophages twice as efficiently as other strains. Macrophage pretreatment with idInlBCC1 followed by recombinant L. monocytogenes infection disturbed macrophage functions decreasing pathogen uptake and improving its intracellular multiplication. Similar pretreatment with idInlBCC7 decreased bacterial uptake but also impaired intracellular multiplication. The obtained results demonstrated that InlB impaired macrophage functions in an idInlB isoform-dependent manner. These data suggest a novel InlB function in L. monocytogenes virulence.


Asunto(s)
Listeria monocytogenes , Listeria , Listeriosis , Humanos , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Isoformas de Proteínas/metabolismo , Factores de Virulencia/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo
5.
Cell Mol Life Sci ; 78(23): 7133-7144, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34626205

RESUMEN

The TLQP-21 neuropeptide has been implicated in functions as diverse as lipolysis, neurodegeneration and metabolism, thus suggesting an important role in several human diseases. Three binding targets have been proposed for TLQP-21: C3aR1, gC1qR and HSPA8. The aim of this review is to critically evaluate the molecular identity of the TLQP-21 receptor and the proposed multi-receptor mechanism of action. Several studies confirm a critical role for C3aR1 in TLQP-21 biological activity and a largely conserved mode of binding, receptor activation and signaling with C3a, its first-identified endogenous ligand. Conversely, data supporting a role of gC1qR and HSPA8 in TLQP-21 activity remain limited, with no signal transduction pathways being described. Overall, C3aR1 is the only receptor for which a necessary and sufficient role in TLQP-21 activity has been confirmed thus far. This conclusion calls into question the validity of a multi-receptor mechanism of action for TLQP-21 and should inform future studies.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas Mitocondriales/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores de Complemento/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Fragmentos de Péptidos/genética , Transducción de Señal/fisiología
6.
Adv Exp Med Biol ; 1295: 29-48, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33543454

RESUMEN

Tumor-homing peptides are widely used for improving tumor selectivity of anticancer drugs and imaging agents. The goal is to increase tumor uptake and reduce accumulation at nontarget sites. Here, we describe current approaches for tumor-homing peptide identification and validation, and provide comprehensive overview of classes of tumor-homing peptides undergoing preclinical and clinical development. We focus on unique mechanistic features and applications of a recently discovered class of tumor-homing peptides, tumor-penetrating C-end Rule (CendR) peptides, that can be used for tissue penetrative targeting of extravascular tumor tissue. Finally, we discuss unanswered questions and future directions in the field of development of peptide-guided smart drugs and imaging agents.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Neoplasias/tratamiento farmacológico , Péptidos/uso terapéutico
7.
Dokl Biochem Biophys ; 500(1): 376-384, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34697746

RESUMEN

The globular heads of the C1q receptor (gC1qR), located in the B cell cytoplasm, perform important roles in many cellular processes. A recent studies reported a major role of mitochondrial apoptosis in several cancers, but there has been no report on gastric carcinoma (GC). In this study, the mechanism by which cell apoptosis is induced by gC1qR in GC was explored. Western blot showed that gC1qR and P53 protein levels were lower in GC tissues than in normal tissues. Cytotoxicity was dynamically increased in gC1qR-overexpressing GC cells compared to the control. CCK8 assay indicated that overexpression of gC1qR induced GC cell apoptosis, increased reactive oxygen species (ROS) production, decreased the mitochondrial transmembrane potential and promoted mitochondrial apoptosis. Moreover, the P53 level increased in response to gC1qR. The viability, migration, and mitochondrial transmembrane potential of GC cells increased in association with decreased levels of ROS and mitochondrial apoptosis in the P53-silenced group. Collectively, our findings indicate that apoptosis of GC cells is enhanced when gC1qR overexpression is induced by P53-mediated mitochondrial apoptosis.


Asunto(s)
Glicoproteínas de Membrana , Receptores de Complemento
8.
BMC Microbiol ; 20(1): 274, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32878596

RESUMEN

BACKGROUND: Clostridioides (formerly Clostridium) difficile infection is the leading cause of antibiotic-associated colitis. Studies have demonstrated that C. difficile toxin A (TcdA) can cause apoptosis of many human cell types. The purpose of this study was to investigate the relationships among exposure to TcdA, the role of the receptor for the globular heads of C1q (gC1qR) gene and the underlying intracellular apoptotic mechanism in human colonic epithelial cells (NCM 460). In this study, gC1qR expression was examined using real-time polymerase chain reaction (PCR), western blotting and immunohistochemical staining. Cell viability was assessed by the water-soluble tetrazolium salt (WST-1) assay, and cell apoptosis was assessed by flow cytometry and the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) assay. Mitochondrial function was assessed based on reactive oxygen species (ROS) generation, changes in the mitochondrial membrane potential (ΔΨm) and the content of ATP. RESULTS: Our study demonstrated that increasing the concentration of TcdA from 10 ng/ml to 20 ng/ml inhibited cell viability and induced cell apoptosis (p < 0.01). Moreover, the TcdA-induced gC1qR expression and enhanced expression of gC1qR caused mitochondrial dysfunction (including production of ROS and decreases in the ΔΨm and the content of ATP) and cell apoptosis. However, silencing of the gC1qR gene reversed TcdA-induced cell apoptosis and mitochondrial dysfunction. CONCLUSION: These data support a mechanism by which gC1qR plays a crucial role in TcdA-induced apoptosis of human colonic epithelial cells in a mitochondria-dependent manner.


Asunto(s)
Apoptosis/efectos de los fármacos , Toxinas Bacterianas/toxicidad , Colon/citología , Enterotoxinas/toxicidad , Glicoproteínas de Membrana/metabolismo , Mitocondrias/efectos de los fármacos , Receptores de Complemento/metabolismo , Adenosina Trifosfato/metabolismo , Apoptosis/fisiología , Línea Celular , Colon/patología , Células Epiteliales , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Complemento/química , Receptores de Complemento/genética
9.
Immunol Rev ; 274(1): 281-289, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27782339

RESUMEN

The blood plasma contains four biologically important proteolytic cascades, which probably evolved from the same ancestral gene. This in part may explain why each cascade has very similar "initiating trigger" followed by sequential and cascade-like downstream enzymatic activation pattern. The four cascades are: the complement system, the blood clotting cascade, the fibrinolytic system, and the kallikrein-kinin system. Although much has been written about the interplay between all these enzymatic cascades, the cross-talk between the complement and the kinin generating systems has become particularly relevant as this interaction results in the generation of nascent molecules that have significant impact in various inflammatory diseases including angioedema and cancer. In this review, we will focus on the consequences of the interplay between the two systems by highlighting the role of a novel molecular link called gC1qR. Although this protein was first identified as a receptor for C1q, it is now recognized as a multiligand binding cellular protein, which serves not only as C1q receptor, but also as high affinity (KD  ≤ 0.8 nM) binding site for both high molecular weight kininogen (HK) and factor XII (FXII). At inflammatory sites, where atherogenic factors such as immune complexes and/or pathogens can activate the endothelial cell into a procoagulant and proinflammatory surface, the two pathways are activated to generate vasoactive peptides that contribute in various ways to the inflammatory processes associated with numerous diseases. More importantly, since recent observations strongly suggest an important role for both pathways in cancer, we will focus on how a growing tumor cluster can employ the byproducts derived from the two activation systems to ensure not only its survival and growth, but also its escape into distal sites of colonization.


Asunto(s)
Angioedema/inmunología , Aterosclerosis/inmunología , Complemento C1q/metabolismo , Factor XIa/metabolismo , Inmunidad Innata , Inflamación/inmunología , Neoplasias/inmunología , Animales , Humanos , Proteolisis , Receptor Cross-Talk , Transducción de Señal
10.
Fish Shellfish Immunol ; 84: 970-978, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30395995

RESUMEN

The receptor for the globular head of complement component C1q, gC1qR, is a multifunctional and multiligand binding protein with a crucial role in host defense. In the present study, a full-length cDNA sequence of a gC1qR homolog (PtgC1qR) in Portunus trituberculatus was identified. PtgC1qR was a 268-amino-acid polypeptide with a conserved MAM33 domain and a mitochondrial targeting sequence in the first 56 amino acids. The transcripts of PtgC1qR were detected in all examined tissues with the highest level detected in the hepatopancreas. Compared with other early embryonic stages, PtgC1qR was highly expressed in the fertilized eggs and embryos at the cleavage stage, which suggest PtgC1qR may be a maternal gene. The transcripts of PtgC1qR in hemocytes exhibited time-dependent response expression pattern after challenged with bacteria (Vibrio alginolyticus, Micrococcus luteus) and fungi (Pichia pastoris). Moreover, the recombinant PtgC1qR (rPtgC1qR) exhibited strong antibacterial activity and microbial-binding activity, suggesting its crucial role in immune defense and recognition. Further phenoloxidase (PO) assay showed that rPtgC1qR could suppress the crab PO activity in vitro in a dose-dependent manner, and it could result in nearly 100% inhibition of PO activity under the concentration of 11.65 µM. Knockdown of PtgC1qR could significantly enhance the expression of serine protease related genes (PtSP1-3 and PtSPH), proPO-associated genes (PtproPO and PtPPAF) and C3-like genes (Ptα2M1 and PtTEP). However, the phagocytosis related genes (PtMyosin, PtRab5 and PtArp) and Ptα2M2 were significantly down-regulated in the PtgC1qR silenced crabs. These findings together demonstrate that PtgC1qR might function in crab immune response via its antibacterial activity, immune recognition or regulating the proPO system, complement pathway and phagocytosis.


Asunto(s)
Braquiuros/genética , Braquiuros/inmunología , Complemento C1q/genética , Complemento C1q/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/inmunología , Secuencia de Bases , Complemento C1q/química , Perfilación de la Expresión Génica , Micrococcus luteus/fisiología , Filogenia , Pichia/fisiología , Vibrio alginolyticus/fisiología
11.
Curr Rheumatol Rep ; 21(11): 60, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31741077

RESUMEN

PURPOSE OF THE REVIEW: Cryoglobulins are immunoglobulins with the ability to precipitate at temperatures <37 °C. They are related to hematological disorders, infections [especially hepatitis C virus (HCV)], and autoimmune diseases. In this article, the state of the art on Cryoglobulinemic Vasculitis (CV), in a helpful and schematic way, with a special focus on HCV related Mixed Cryoglobulinemia treatment are reviewed. RECENT FINDINGS: Direct - acting antivirals (DAA) against HCV have emerged as an important key in HCV treatment to related Cryoglobulinemic Vasculitis, and should be kept in mind as the initial treatment in non-severe manifestations. On the other hand, a recent consensus panel has published their recommendations for treatment in severe and life threatening manifestations of Mixed Cryoglobulinemias. HCV-Cryoglobulinemic vasculitis is the most frequent form of CV. There are new treatment options in HCV-CV with DAA, with an important number of patients achieving complete response and sustained virologic response (SVR). In cases of severe forms of CV, treatment with Rituximab and PLEX are options. The lack of data on maintenance therapy could impulse future studies in this setting.


Asunto(s)
Antivirales/uso terapéutico , Vasculitis/tratamiento farmacológico , Crioglobulinemia/diagnóstico , Crioglobulinemia/tratamiento farmacológico , Humanos , Resultado del Tratamiento , Vasculitis/diagnóstico
12.
Biochem J ; 475(4): 775-786, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29358188

RESUMEN

The protein gC1qR (globular C1q receptor), also named p33, was originally identified as a binding partner of the globular heads of C1q in the complement system. gC1qR/p33 is abundantly expressed in many cell types, but the functional importance of this protein is not completely understood. Here, we investigate the impact of gC1qR/p33 on the production and function of the pathophysiologically important chemokine monocyte chemoattractant protein-1 (MCP-1) and the underlying molecular mechanisms. Knockdown of gC1qR/p33 negatively regulated the production of MCP-1, but had no effect on the expression of transcript for MCP-1 in human periodontal ligament cells, suggesting a translational/post-translational mechanism of action. Laser scanning confocal microscopy showed considerable cytosolic co-localization of gC1qR/p33 and MCP-1, and co-immunoprecipitation disclosed direct physical interaction between gC1qR/p33 and MCP-1. Surface plasmon resonance analysis revealed a high-affinity binding (KD = 10.9 nM) between gC1qR/p33 and MCP-1. Using a transwell migration assay, we found that recombinant gC1qR/p33 enhances MCP-1-induced migration of human THP-1 monocytes, pointing to a functional importance of the interaction between gC1qR/p33 and MCP-1. An in vitro assay revealed a rapid turnover of the MCP-1 protein and that gC1qR/p33 stabilizes MCP-1, hence preventing its degradation. We propose that endogenous gC1qR/p33 physically interacts with MCP-1 causing stabilization of the MCP-1 protein and stimulation of its activity in human periodontal ligament cells, suggesting a novel gC1qR/p33-mediated pro-inflammatory mechanism of action.


Asunto(s)
Proteínas Portadoras/genética , Quimiocina CCL2/genética , Inflamación/genética , Proteínas Mitocondriales/genética , Ligamento Periodontal/metabolismo , Proteínas Portadoras/metabolismo , Movimiento Celular/genética , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/química , Citosol/química , Citosol/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo , Inflamación/patología , Microscopía Confocal , Proteínas Mitocondriales/metabolismo , Monocitos/metabolismo , Monocitos/patología , Ligamento Periodontal/crecimiento & desarrollo , Ligamento Periodontal/patología , Unión Proteica , Procesamiento Proteico-Postraduccional/genética , Resonancia por Plasmón de Superficie
13.
Int J Mol Sci ; 20(17)2019 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-31450632

RESUMEN

The pathogenic Gram-positive bacterium Listeria monocytogenes has been evolving into a few phylogenetic lineages. Phylogenetically defined substitutions were described in the L. monocytogenes virulence factor InlB, which mediates active invasion into mammalian cells via interactions with surface receptors c-Met and gC1q-R. InlB internalin domain (idInlB) is central to interactions with c-Met. Here we compared activity of purified recombinant idInlB isoforms characteristic for L. monocytogenes phylogenetic lineage I and II. Size exclusion chromatography and intrinsic fluorescence were used to characterize idInlBs. Western blotting was used to study activation of c-Met-dependent MAPK- and PI3K/Akt-pathways. Solid-phase microplate binding and competition assay was used to quantify interactions with gCq1-R. Isogenic recombinant L. monocytogenes strains were used to elucidate the input of idInlB isoforms in HEp-2 cell invasion. Physicochemical parameters of idInlB isoforms were similar but not identical. Kinetics of Erk1/2 and Akt phosphorylation in response to purified idInlBs was lineage specific. Lineage I but not lineage II idInlB specifically bound gC1q-R. Antibody against gC1q-R amino acids 221-249 inhibited invasion of L. monocytogenes carrying lineage I but not lineage II idInlB. Taken together, obtained results suggested that phylogenetically defined substitutions in idInlB provide functional distinctions and might be involved in phylogenetically determined differences in virulence potential.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Listeria monocytogenes/clasificación , Listeria monocytogenes/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Filogenia , Transducción de Señal , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Línea Celular , Humanos , Listeria monocytogenes/patogenicidad , Proteínas de la Membrana/química , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas , Factores de Virulencia
14.
Biochem Biophys Res Commun ; 489(4): 460-465, 2017 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-28576489

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of respiratory infections in infants and the elderly, leading to more deaths than influenza each year, but there is no antiviral or efficacious vaccine currently available. Here we examine the role in infection of the host mitochondrial protein p32 (HABP/gC1qR/C1qbp) for the first time. RSV replication as well as infectious virus production was significantly reduced by p32 siRNA knockdown, consistent with an important role for p32 in RSV infection. p32 showed distinct mitochondrial localization throughout RSV infection, but immunostaining and high resolution confocal imaging for p32 as well as MitoTracker Red and cytochrome c, revealed clear changes in mitochondrial organization in RSV infection, with perinuclear mitochondrial compaction and asymmetric distribution at 8 and 18 h post-infection, respectively. The results implicate p32 as a key host factor for RSV virus production, and bring to light the potential importance of mitochondria in RSV infection.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Mitocondriales/metabolismo , Virus Sincitiales Respiratorios/crecimiento & desarrollo , Virus Sincitiales Respiratorios/metabolismo , Células A549 , Proteínas Portadoras/genética , Humanos , Proteínas Mitocondriales/genética , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas
15.
Biochem J ; 473(1): 87-98, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26508735

RESUMEN

The human host-defence peptide (HDP) LL-37 not only displays anti-microbial activity but also immune-modulating properties that trigger intracellular signalling events in host cells. Since the cytolytic activity of high LL-37 concentrations affects cell viability, the function of LL-37 requires tight regulation. Eukaryotic cells therefore benefit from protective measures to prevent harmful effects of LL-37. p33, also known as globular C1q receptor (gC1qR), is reported to act as an LL-37 antagonist by binding the peptide, thereby reducing its cytotoxic activity. In the present report, we show that high levels of endogenous p33 correlate with an increased viability in human cells treated with LL-37. Sub-cellular localization analysis showed p33 distribution at the mitochondria, the plasma membrane and in the cytosol. Strikingly, cytosolic overexpression of p33 significantly antagonized detrimental effects of LL-37 on cell fitness, whereas the reverse effect was observed by siRNA-induced down-regulation of p33. However, modulation of p33 expression had no effect on LL-37-induced plasma membrane pore forming capacity pointing to an intracellular mechanism. A scavenging function of intracellular p33 is further supported by co-immunoprecipitation experiments, showing a direct interaction between intracellular p33 and LL-37. Thus, our findings support an important role of intracellular p33 in maintaining cell viability by counteracting LL-37-induced cytotoxicity.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/toxicidad , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Citotoxinas/toxicidad , Glicoproteínas de Membrana/biosíntesis , Receptores de Complemento/biosíntesis , Adolescente , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica , Células HeLa , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Receptores de Complemento/genética , Catelicidinas
16.
J Biol Chem ; 289(52): 35795-805, 2014 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-25355318

RESUMEN

As a large double-stranded DNA virus, herpes simplex virus type 1 (HSV-1) assembles capsids in the nucleus where the viral particles exit by budding through the inner nuclear membrane. Although a number of viral and host proteins are involved, the machinery of viral egress is not well understood. In a search for host interacting proteins of ICP34.5, which is a virulence factor of HSV-1, we identified a cellular protein, p32 (gC1qR/HABP1), by mass spectrophotometer analysis. When expressed, ICP34.5 associated with p32 in mammalian cells. Upon HSV-1 infection, p32 was recruited to the inner nuclear membrane by ICP34.5, which paralleled the phosphorylation and rearrangement of nuclear lamina. Knockdown of p32 in HSV-1-infected cells significantly reduced the production of cell-free viruses, suggesting that p32 is a mediator of HSV-1 nuclear egress. These observations suggest that the interaction between HSV-1 ICP34.5 and p32 leads to the disintegration of nuclear lamina and facilitates the nuclear egress of HSV-1 particles.


Asunto(s)
Proteínas Portadoras/metabolismo , Núcleo Celular/virología , Herpesvirus Humano 1/fisiología , Proteínas Mitocondriales/metabolismo , Proteínas Virales/metabolismo , Animales , Chlorocebus aethiops , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Receptores Citoplasmáticos y Nucleares/metabolismo , Células Vero , Liberación del Virus , Receptor de Lamina B
17.
J Biol Chem ; 288(48): 34638-46, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24106277

RESUMEN

VGF (nonacronymic) is a neuropeptide precursor that plays multiple roles in regulation of energy balance, reproduction, hippocampal synaptic plasticity, and pain. Data from a number of pain models showed significant up-regulation of VGF in sensory neurons. TLQP-21, one of the VGF-derived neuropeptides, has been shown to induce a hyperalgesic response when injected subcutaneously into the hind paw of mice. However, the precise role of VGF-derived neuropeptides in neuropathic pain and the molecular identity of the receptor for VGF-derived peptides are yet to be investigated. Here we identified gC1qR, the globular heads of the C1q receptor, as the receptor for TLQP-21 using chemical cross-linking combined with mass spectrometry analysis. TLQP-21 caused an increase in intracellular Ca(2+) levels in rat macrophages and microglia. Inoculation of TLQP-21-stimulated macrophages into rat hind paw caused mechanical hypersensitivity. The increase in intracellular Ca(2+) levels in macrophages was attenuated by either siRNA or neutralizing antibodies against gC1qR. Furthermore, application of the gC1qR-neutralizing antibody to rats with partial sciatic nerve ligation resulted in a delayed onset of nerve injury-associated mechanical hypersensitivity. These results indicate that gC1qR is the receptor for TLQP-21 and plays an important role in chronic pain through activation of macrophages. Because direct association between TLQP-21 and gC1qR is required for activation of macrophages and causes hypersensitivity, disrupting this interaction may be a useful new approach to develop novel analgesics.


Asunto(s)
Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Neuralgia/metabolismo , Neuropéptidos/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores de Complemento/metabolismo , Animales , Calcio/metabolismo , Humanos , Espectrometría de Masas , Ratones , Microglía/metabolismo , Neuralgia/patología , Fragmentos de Péptidos/administración & dosificación , Ratas , Receptores de Neuropéptido/metabolismo , Células Receptoras Sensoriales/metabolismo
18.
Front Allergy ; 5: 1302605, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38332896

RESUMEN

Anaphylaxis is a potentially life-threatening multi-system allergic reaction to a biological trigger resulting in the release of potent inflammatory mediators from mast cells and basophils and causing symptoms in at least two organ systems that generally include skin, lungs, heart, or gastrointestinal tract in any combination. One exception is profound hypotension as an isolated symptom. There are two types of triggers of anaphylaxis: immunologic and non-Immunologic. Immunologic anaphylaxis is initiated when a foreign antigen directly binds to IgE expressed on mast cells or basophils and induces the release of histamine and other inflammatory substances resulting in vasodilation, vascular leakage, decreased peripheral vascular resistance, and heart muscle depression. If left untreated, death by shock (profound hypotension) or asphyxiation (airway obstruction) can occur. The non-immunologic pathway, on the other hand, can be initiated in many ways. A foreign substance can directly bind to receptors of mast cells and basophils leading to degranulation. There can be immune complex activation of the classical complement cascade with the release of anaphylatoxins C3a and C5a with subsequent recruitment of mast cells and basophils. Finally, hyperosmolar contrast agents can cause blood cell lysis, enzyme release, and complement activation, resulting in anaphylactoid (anaphylactic-like) symptoms. In this report we emphasize the recruitment of the bradykinin-forming cascade in mast cell dependent anaphylactic reactions as a potential mediator of severe hypotension, or airway compromise (asthma, laryngeal edema). We also consider airway obstruction due to inhibition of angiotensin converting enzyme with a diminished rate of endogenous bradykinin metabolism, leading not only to laryngeal edema, but massive tongue swelling with aspiration of secretions.

19.
Front Immunol ; 15: 1405597, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38983846

RESUMEN

Endometriosis (EM) is defined as the engraftment and proliferation of functional endometrial-like tissue outside the uterine cavity, leading to a chronic inflammatory condition. While the precise etiology of EM remains elusive, recent studies have highlighted the crucial involvement of a dysregulated immune system. The complement system is one of the predominantly altered immune pathways in EM. Owing to its involvement in the process of angiogenesis, here, we have examined the possible role of the first recognition molecule of the complement classical pathway, C1q. C1q plays seminal roles in several physiological and pathological processes independent of complement activation, including tumor growth, placentation, wound healing, and angiogenesis. Gene expression analysis using the publicly available data revealed that C1q is expressed at higher levels in EM lesions compared to their healthy counterparts. Immunohistochemical analysis confirmed the presence of C1q protein, being localized around the blood vessels in the EM lesions. CD68+ macrophages are the likely producer of C1q in the EM lesions since cultured EM cells did not produce C1q in vitro. To explore the underlying reasons for increased C1q expression in EM, we focused on its established pro-angiogenic role. Employing various angiogenesis assays on primary endothelial endometriotic cells, such as migration, proliferation, and tube formation assays, we observed a robust proangiogenic effect induced by C1q on endothelial cells in the context of EM. C1q promoted angiogenesis in endothelial cells isolated from EM lesions (as well as healthy ovary that is also rich in C1q). Interestingly, endothelial cells from EM lesions seem to overexpress the receptor for the globular heads of C1q (gC1qR), a putative C1q receptor. Experiments with siRNA to silence gC1qR resulted in diminished capacity of C1q to perform its angiogenic functions, suggesting that C1q is likely to engage gC1qR in the pathophysiology of EM. gC1qR can be a potential therapeutic target in EM patients that will disrupt C1q-mediated proangiogenic activities in EM.


Asunto(s)
Complemento C1q , Endometriosis , Neovascularización Patológica , Endometriosis/metabolismo , Endometriosis/inmunología , Endometriosis/patología , Endometriosis/genética , Complemento C1q/genética , Complemento C1q/metabolismo , Humanos , Femenino , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/inmunología , Endometrio/inmunología , Endometrio/metabolismo , Endometrio/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Células Cultivadas , Adulto , Proliferación Celular
20.
Front Immunol ; 15: 1351656, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38711524

RESUMEN

Understanding at the molecular level of the cell biology of tumors has led to significant treatment advances in the past. Despite such advances however, development of therapy resistance and tumor recurrence are still unresolved major challenges. This therefore underscores the need to identify novel tumor targets and develop corresponding therapies to supplement existing biologic and cytotoxic approaches so that a deeper and more sustained treatment responses could be achieved. The complement system is emerging as a potential novel target for cancer therapy. Data accumulated to date show that complement proteins, and in particular C1q and its receptors cC1qR/CR and gC1qR/p33/HABP1, are overexpressed in most cancer cells and together are involved not only in shaping the inflammatory tumor microenvironment, but also in the regulation of angiogenesis, metastasis, and cell proliferation. In addition to the soluble form of C1q that is found in plasma, the C1q molecule is also found anchored on the cell membrane of monocytes, macrophages, dendritic cells, and cancer cells, via a 22aa long leader peptide found only in the A-chain. This orientation leaves its 6 globular heads exposed outwardly and thus available for high affinity binding to a wide range of molecular ligands that enhance tumor cell survival, migration, and proliferation. Similarly, the gC1qR molecule is not only overexpressed in most cancer types but is also released into the microenvironment where it has been shown to be associated with cancer cell proliferation and metastasis by activation of the complement and kinin systems. Co-culture of either T cells or cancer cells with purified C1q or anti-gC1qR has been shown to induce an anti-proliferative response. It is therefore postulated that in the tumor microenvironment, the interaction between C1q expressing cancer cells and gC1qR bearing cytotoxic T cells results in T cell suppression in a manner akin to the PD-L1 and PD-1 interaction.


Asunto(s)
Proteínas Portadoras , Complemento C1q , Inhibidores de Puntos de Control Inmunológico , Glicoproteínas de Membrana , Proteínas Mitocondriales , Neoplasias , Receptores de Complemento , Humanos , Complemento C1q/metabolismo , Complemento C1q/inmunología , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Receptores de Complemento/metabolismo , Animales , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA