Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 500
Filtrar
1.
Cancer Res Commun ; 4(2): 505-515, 2024 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-38319147

RESUMO

In normal cells, binding of the transmembrane protein CD47 to signal regulatory protein-α (SIRPα) on macrophages induces an antiphagocytic signal. Tumor cells hijack this pathway and overexpress CD47 to evade immune destruction. Macrophage antitumor activity can be restored by simultaneously blocking the CD47-SIRPα signaling axis and inducing a prophagocytic signal via tumor-opsonizing antibodies. We identified a novel, fully human mAb (BMS-986351) that binds SIRPα with high affinity. BMS-986351 demonstrated broad binding coverage across SIRPα polymorphisms and potently blocked CD47-SIRPα binding at the CD47 binding site in a dose-dependent manner. In vitro, BMS-986351 increased phagocytic activity against cell lines from solid tumors and hematologic malignancies, and this effect was markedly enhanced when BMS-986351 was combined with the opsonizing antibodies cetuximab and rituximab. A phase I dose-escalation/-expansion study of BMS-986351 for the treatment of advanced solid and hematologic malignancies is underway (NCT03783403). SIGNIFICANCE: Increasing the phagocytotic capabilities of tumor-associated macrophages by modulating macrophage-tumor cell surface signaling via the CD47-SIRPα axis is a novel strategy. Molecules targeting CD47 have potential but its ubiquitous expression necessitates higher therapeutic doses to overcome potential antigen sink effects. The restricted expression pattern of SIRPα may limit toxicities and lower doses of the SIRPα antibody BMS-986351 may overcome target mediated drug disposition while maintaining the desired pharmacology.


Assuntos
Neoplasias Hematológicas , Neoplasias , Humanos , Antígeno CD47/genética , Receptores Imunológicos/genética , Fagocitose , Macrófagos , Neoplasias/tratamento farmacológico , Anticorpos Antineoplásicos/metabolismo , Proteínas Opsonizantes/metabolismo , Neoplasias Hematológicas/metabolismo
2.
Cell Biochem Funct ; 41(8): 1031-1043, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37933222

RESUMO

Opsonization plays a pivotal role in hindering controlled drug release from nanoformulations due to macrophage-mediated nanoparticle destruction. While first and second-generation delivery systems, such as lipoplexes (50-150 nm) and quantum dots, hold immense potential in revolutionizing disease treatment through spatiotemporal controlled drug delivery, their therapeutic efficacy is restricted by the selective labeling of nanoparticles for uptake by reticuloendothelial system and mononuclear phagocyte system via various molecular forces, such as electrostatic, hydrophobic, and van der Waals bonds. This review article presents novel insights into surface-modification techniques utilizing macromolecule-mediated approaches, including PEGylation, di-block copolymerization, and multi-block polymerization. These techniques induce stealth properties by generating steric forces to repel micromolecular-opsonins, such as fibrinogen, thereby mitigating opsonization effects. Moreover, advanced biological methods, like cellular hitchhiking and dysopsonic protein adsorption, are highlighted for their potential to induce biological camouflage by adsorbing onto the nanoparticulate surface, leading to immune escape. These significant findings pave the way for the development of long-circulating next-generation nanoplatforms capable of delivering superior therapy to patients. Future integration of artificial intelligence-based algorithms, integrated with nanoparticle properties such as shape, size, and surface chemistry, can aid in elucidating nanoparticulate-surface morphology and predicting interactions with the immune system, providing valuable insights into the probable path of opsonization.


Assuntos
Nanopartículas , Polietilenoglicóis , Humanos , Polietilenoglicóis/química , Opsonização , Inteligência Artificial , Sistemas de Liberação de Medicamentos , Proteínas Opsonizantes/química , Proteínas Opsonizantes/metabolismo , Nanopartículas/química
3.
Front Immunol ; 14: 1238411, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37860004

RESUMO

C-reactive protein (CRP) is one of the major members of the family of acute phase proteins (APP). Interest in this CRP was the result of a seminal discovery of its pattern of response to pneumococcal infection in humans. CRP has the unique property of reacting with phosphocholine-containing substances, such as pneumococcal C-polysaccharide, in the presence of Ca2+. The attention regarding the origin of CRP and its multifunctionality has gripped researchers for several decades. The reason can be traced to the integrated evolution of CRP in the animal kingdom. CRP has been unequivocally listed as a key indicator of infectious and inflammatory diseases including autoimmune diseases. The first occurrence of CRP in the evolutionary ladder appeared in arthropods followed by molluscs and much later in the chordates. The biological significance of CRP has been established in the animal kingdom starting from invertebrates. Interestingly, the site of synthesis of CRP is mainly the liver in vertebrates, while in invertebrates it is located in diverse tissues. CRP is a multifunctional player in the scenario of innate immunity. CRP acts as an opsonin in the area of complement activation and phagocytosis. Interestingly, CRP upregulates and downregulates both cytokine production and chemotaxis. Considering various studies of CRP in humans and non-human animals, it has been logically proposed that CRP plays a common role in animals. CRP also interacts with Fcγ receptors and triggers the inflammatory response of macrophages. CRP in other animals such as primates, fish, echinoderms, arthropods, and molluscs has also been studied in some detail which establishes the evolutionary significance of CRP. In mammals, the increase in CRP levels is an induced response to inflammation or trauma; interestingly, in arthropods and molluscs, CRP is constitutively expressed and represents a major component of their hemolymph. Investigations into the primary structure of CRP from various species revealed the overall relatedness between vertebrate and invertebrate CRP. Invertebrates lack an acquired immune response; they are therefore dependent on the multifunctional role of CRP leading to the evolutionary success of the invertebrate phyla.


Assuntos
Proteína C-Reativa , Inflamação , Animais , Proteína C-Reativa/metabolismo , Invertebrados , Mamíferos , Proteínas Opsonizantes/metabolismo , Fagocitose , Humanos
4.
Front Immunol ; 13: 981375, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36189215

RESUMO

The complement system is part of the innate immune system. The crucial step in activating the complement system is the generation and regulation of C3 convertase complexes, which are needed to generate opsonins that promote phagocytosis, to generate C3a that regulates inflammation, and to initiate the lytic terminal pathway through the generation and activity of C5 convertases. A growing body of evidence has highlighted the interplay between the complement system, coagulation system, platelets, neutrophils, and endothelial cells. The kidneys are highly susceptible to complement-mediated injury in several genetic, infectious, and autoimmune diseases. Atypical hemolytic uremic syndrome (aHUS) and lupus nephritis (LN) are both characterized by thrombosis in the glomerular capillaries of the kidneys. In aHUS, congenital or acquired defects in complement regulators may trigger platelet aggregation and activation, resulting in the formation of platelet-rich thrombi in the kidneys. Because glomerular vasculopathy is usually noted with immunoglobulin and complement accumulation in LN, complement-mediated activation of tissue factors could partly explain the autoimmune mechanism of thrombosis. Thus, kidney glomerular capillary thrombosis is mediated by complement dysregulation and may also be associated with complement overactivation. Further investigation is required to clarify the interaction between these vascular components and develop specific therapeutic approaches.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Trombose , Síndrome Hemolítico-Urêmica Atípica/genética , Capilares/metabolismo , Convertases de Complemento C3-C5/metabolismo , Proteínas do Sistema Complemento/metabolismo , Células Endoteliais/metabolismo , Humanos , Rim/metabolismo , Proteínas Opsonizantes/metabolismo , Trombose/metabolismo
5.
Small ; 18(40): e2203746, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36070419

RESUMO

Bloodstream infection caused by antimicrobial resistance pathogens is a global concern because it is difficult to treat with conventional therapy. Here, scavenger magnetic nanoparticles enveloped by nanovesicles derived from blood cells (MNVs) are reported, which magnetically eradicate an extreme range of pathogens in an extracorporeal circuit. It is quantitatively revealed that glycophorin A and complement receptor (CR) 1 on red blood cell (RBC)-MNVs predominantly capture human fecal bacteria, carbapenem-resistant (CR) Escherichia  coli, and extended-spectrum beta-lactamases-positive (ESBL-positive) E. coli, vancomycin-intermediate Staphylococcus aureus (VISA), endotoxins, and proinflammatory cytokines in human blood. Additionally, CR3 and CR1 on white blood cell-MNVs mainly contribute to depleting the virus envelope proteins of Zika, SARS-CoV-2, and their variants in human blood. Supplementing opsonins into the blood significantly augments the pathogen removal efficiency due to its combinatorial interactions between pathogens and CR1 and CR3 on MNVs. The extracorporeal blood cleansing enables full recovery of lethally infected rodent animals within 7 days by treating them twice in series. It is also validated that parameters reflecting immune homeostasis, such as blood cell counts, cytokine levels, and transcriptomics changes, are restored in blood of the fatally infected rats after treatment.


Assuntos
Bacteriemia , Tratamento Farmacológico da COVID-19 , Infecções por Escherichia coli , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bacteriemia/tratamento farmacológico , Bacteriemia/microbiologia , Carbapenêmicos/metabolismo , Citocinas/metabolismo , Endotoxinas/metabolismo , Escherichia coli/metabolismo , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/microbiologia , Glicoforinas/metabolismo , Homeostase , Humanos , Testes de Sensibilidade Microbiana , Proteínas Opsonizantes/metabolismo , Ratos , Receptores de Complemento/metabolismo , Roedores/metabolismo , SARS-CoV-2 , Proteínas do Envelope Viral/metabolismo , beta-Lactamases/metabolismo
6.
J Biol Chem ; 298(9): 102269, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35850306

RESUMO

Carcinoembryonic antigen-related cell adhesion molecule 3 (CEACAM3) is a human granulocyte receptor mediating the efficient phagocytosis of a subset of human-restricted bacterial pathogens. Its function depends on phosphorylation of a tyrosine-based sequence motif, but the enzyme(s) responsible for reversing this modification are unclear. Here, we identify the receptor-type protein tyrosine phosphatase PTPRJ as a negative regulator of CEACAM3-mediated phagocytosis. We show depletion of PTPRJ results in a gain-of-function phenotype, while overexpression of a constitutively active PTPRJ phosphatase strongly reduces bacterial uptake via CEACAM3. We also determined that recombinant PTPRJ directly dephosphorylates the cytoplasmic tyrosine residues of purified full-length CEACAM3 and recognizes synthetic CEACAM3-derived phosphopeptides as substrates. Dephosphorylation of CEACAM3 by PTPRJ is also observed in intact cells, thereby limiting receptor-initiated cytoskeletal re-arrangements, lamellipodia formation, and bacterial uptake. Finally, we show that human phagocytes deficient for PTPRJ exhibit exaggerated lamellipodia formation and enhanced opsonin-independent phagocytosis of CEACAM3-binding bacteria. Taken together, our results highlight PTPRJ as a bona fide negative regulator of CEACAM3-initiated phagocyte functions, revealing a potential molecular target to limit CEACAM3-driven inflammatory responses.


Assuntos
Antígeno Carcinoembrionário , Fagocitose , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores , Antígeno Carcinoembrionário/metabolismo , Granulócitos/metabolismo , Humanos , Proteínas Opsonizantes/metabolismo , Fagocitose/fisiologia , Fosfopeptídeos/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo
7.
Adv Mater ; 34(34): e2203477, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35793262

RESUMO

Real-time monitoring of neutrophil dynamics is crucial for timely diagnosis and effective treatment of inflammation-related diseases, which requires a reliable tracer for in vivo tracking of neutrophils. However, immunotracers for neutrophils are extremely limited because of the difficulty in labeling the cells. Inspired by the natural biological function of the complement system, a strategy of enhancing the complement C3 opsonization of lanthanide-doped nanoparticles (LnNPs) by modulating their surface chemistry, thus developing a near infrared-IIb emissive nanotracer for neutrophils, is reported herein. Four kinds of surface-modified LnNPs are fabricated, among which phospholipids DOPG-modified LnNPs (LnNPs@PG) with weak antifouling ability and hydroxyl groups adsorb more complement C3 proteins and form covalent linkages with C3b active fragments under inflammation conditions, inducing enhanced complement C3 opsonization. Therefore, LnNPs@PG with enhanced complement C3 opsonization are capable of efficiently labeling inflammation-stimulated neutrophils in vivo through complement-receptors-mediated phagocytosis and achieve dynamic monitoring neutrophils during cutaneous wound healing and cerebral ischemia/reperfusion.


Assuntos
Complemento C3 , Neutrófilos , Complemento C3/metabolismo , Humanos , Inflamação/metabolismo , Neutrófilos/metabolismo , Proteínas Opsonizantes/metabolismo , Fagocitose
8.
Small ; 18(14): e2106529, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35187804

RESUMO

Coating nanoparticles with poly(ethylene glycol) (PEG) is widely used to achieve long-circulating properties after infusion. While PEG reduces binding of opsonins to the particle surface, immunogenic anti-PEG side-effects show that PEGylated nanoparticles are not truly "stealth" to surface active proteins. A major obstacle for understanding the complex interplay between opsonins and nanoparticles is the averaging effects of the bulk assays that are typically applied to study protein adsorption to nanoparticles. Here, a microscopy-based method for directly quantifying opsonization at the single nanoparticle level is presented. Various surface coatings are investigated on liposomes, including PEG, and show that opsonization by both antibodies and complement C3b is highly dependent on the surface chemistry. It is further demonstrated that this opsonization is heterogeneous, with opsonized and non-opsonized liposomes co-existing in the same ensemble. Surface coatings modify the percentage of opsonized liposomes and/or opsonin surface density on the liposomes, with strikingly different patterns for antibodies and complement. Thus, this assay provides mechanistic details about opsonization at the single nanoparticle level previously inaccessible to established bulk assays.


Assuntos
Lipossomos , Proteínas Opsonizantes , Anticorpos , Proteínas do Sistema Complemento/metabolismo , Lipossomos/química , Proteínas Opsonizantes/metabolismo , Opsonização , Polietilenoglicóis/química
9.
Nat Nanotechnol ; 17(1): 86-97, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34795440

RESUMO

This study shows that the supramolecular arrangement of proteins in nanoparticle structures predicts nanoparticle accumulation in neutrophils in acute lung inflammation (ALI). We observed homing to inflamed lungs for a variety of nanoparticles with agglutinated protein (NAPs), defined by arrangement of protein in or on the nanoparticles via hydrophobic interactions, crosslinking and electrostatic interactions. Nanoparticles with symmetric protein arrangement (for example, viral capsids) had no selectivity for inflamed lungs. Flow cytometry and immunohistochemistry showed NAPs have tropism for pulmonary neutrophils. Protein-conjugated liposomes were engineered to recapitulate NAP tropism for pulmonary neutrophils. NAP uptake in neutrophils was shown to depend on complement opsonization. We demonstrate diagnostic imaging of ALI with NAPs; show NAP tropism for inflamed human donor lungs; and show that NAPs can remediate pulmonary oedema in ALI. This work demonstrates that structure-dependent tropism for neutrophils drives NAPs to inflamed lungs and shows NAPs can detect and treat ALI.


Assuntos
Inflamação/patologia , Pulmão/patologia , Nanopartículas/química , Neutrófilos/patologia , Proteínas/química , Doença Aguda , Aglutinação/efeitos dos fármacos , Animais , Anticorpos/farmacologia , Reagentes de Ligações Cruzadas/química , Dextranos/química , Humanos , Lipopolissacarídeos , Lipossomos , Pulmão/diagnóstico por imagem , Masculino , Camundongos Endogâmicos C57BL , Muramidase/metabolismo , Neutrófilos/efeitos dos fármacos , Proteínas Opsonizantes/metabolismo , Eletricidade Estática , Distribuição Tecidual/efeitos dos fármacos , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
10.
J Immunol Res ; 2021: 2185568, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34765679

RESUMO

We recently showed that both nontypeable Haemophilus influenzae (NTHi) and its surface plasminogen- (Plg-) binding proteins interact with lipoprotein(a) (Lp(a)) in a lysine-dependent manner. Because Lp(a) can be taken up by macrophages, we postulated that it serves as an opsonin to enhance phagocytosis of NTHi by macrophages. Based on colony-forming unit (CFU) counts, Lp(a) was found to increase U937 macrophage-mediated phagocytosis of NTHi49247 and NTHi49766 by 34% and 43%, respectively, after 120 min. In contrast, Lp(a) did not enhance phagocytosis of Escherichia coli BL21 or E. coli JM109, which were unable to bind to Lp(a). As with U937 macrophages, Lp(a) was capable of increasing phagocytosis of NTHi49247 by peripheral blood mononuclear cell-derived macrophages. Opsonic phagocytosis by Lp(a) was inhibited by the addition of recombinant kringle IV type 10 (rKIV10), a lysine-binding competitor; moreover, Lp(a) did not increase phagocytosis of NTHi by U937 macrophages that were pretreated with a monoclonal antibody against the scavenger receptor CD36. Taken together, our observation suggests that Lp(a) might serve as a lysine-binding opsonin to assist macrophages in rapid recognition and phagocytosis of NTHi.


Assuntos
Infecções por Haemophilus/metabolismo , Haemophilus influenzae/patogenicidade , Lipoproteína(a)/metabolismo , Macrófagos/metabolismo , Proteínas Opsonizantes/metabolismo , Fagocitose/fisiologia , Antígenos CD36/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Escherichia coli/patogenicidade , Infecções por Haemophilus/microbiologia , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/microbiologia , Macrófagos/microbiologia , Células U937
11.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-33799879

RESUMO

The development of nanocarriers (NC) for biomedical applications has gained large interest due to their potential to co-deliver drugs in a cell-type-targeting manner. However, depending on their surface characteristics, NC accumulate serum factors, termed protein corona, which may affect their cellular binding. We have previously shown that NC coated with carbohydrates to enable biocompatibility triggered the lectin-dependent complement pathway, resulting in enhanced binding to B cells via complement receptor (CR)1/2. Here we show that such NC also engaged all types of splenic leukocytes known to express CR3 at a high rate when NC were pre-incubated with native mouse serum resulting in complement opsonization. By focusing on dendritic cells (DC) as an important antigen-presenting cell type, we show that CR3 was essential for binding/uptake of complement-opsonized NC, whereas CR4, which in mouse is specifically expressed by DC, played no role. Further, a minor B cell subpopulation (B-1), which is important for first-line pathogen responses, and co-expressed CR1/2 and CR3, in general, engaged NC to a much higher extent than normal B cells. Here, we identified CR-1/2 as necessary for binding of complement-opsonized NC, whereas CR3 was dispensable. Interestingly, the binding of complement-opsonized NC to both DC and B-1 cells affected the expression of activation markers. Our findings may have important implications for the design of nano-vaccines against infectious diseases, which codeliver pathogen-specific protein antigen and adjuvant, aimed to induce a broad adaptive cellular and humoral immune response by inducing cytotoxic T lymphocytes that kill infected cells and pathogen-neutralizing antibodies, respectively. Decoration of nano-vaccines either with carbohydrates to trigger complement activation in vivo or with active complement may result in concomitant targeting of DC and B cells and thereby may strongly enhance the extent of dual cellular/humoral immune responses.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Antígeno CD11b/imunologia , Proteínas do Sistema Complemento/imunologia , Células Dendríticas/imunologia , Receptores de Complemento/imunologia , Animais , Subpopulações de Linfócitos B/metabolismo , Linfócitos B/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Células Cultivadas , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Células Dendríticas/metabolismo , Dextranos/química , Portadores de Fármacos/química , Humanos , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nanopartículas/química , Proteínas Opsonizantes/imunologia , Proteínas Opsonizantes/metabolismo , Fagocitose/imunologia , Receptores de Complemento/metabolismo
12.
Front Immunol ; 12: 635825, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33679791

RESUMO

Neutrophils play a key role in the human immune response to Staphylococcus aureus infections. These professional phagocytes rapidly migrate to the site of infection to engulf bacteria and destroy them via specialized intracellular killing mechanisms. Here we describe a robust and relatively high-throughput flow cytometry assay to quantify phagocytosis of S. aureus by human neutrophils. We show that effective phagocytic uptake of S. aureus is greatly enhanced by opsonization, i.e. the tagging of microbial surfaces with plasma-derived host proteins like antibodies and complement. Our rapid assay to monitor phagocytosis can be used to study neutrophil deficiencies and bacterial evasion, but also provides a powerful tool to assess the opsonic capacity of antibodies, either in the context of natural immune responses or immune therapies.


Assuntos
Técnicas Bacteriológicas , Citometria de Fluxo , Neutrófilos/microbiologia , Fagocitose , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/metabolismo , Células Cultivadas , Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Evasão da Resposta Imune , Neutrófilos/imunologia , Neutrófilos/metabolismo , Proteínas Opsonizantes/imunologia , Proteínas Opsonizantes/metabolismo , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/imunologia , Fatores de Tempo
13.
Dev Comp Immunol ; 120: 104066, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33705791

RESUMO

In the immune system, C-type lectins, as pattern recognition receptors, have an important function. Carbohydrate-recognition domains (CRDs) endow C-type lectins with the function of recognizing and scavenging non-self factors. In the present study, a new C-type lectin (designated as PtCTL-9 according to the order of discovery) from swimming crab (Portunus trituberculatus) was characterized. QPN (Gln-Pro-Asn) and FHS (Phe-His-Ser) were identified as the key motifs that determine carbohydrate binding. Motif QPN was mutated to QPD (Gln-Pro-Asp) (M1) and EPN (Glu-Pro-Asn) (M2) to study its immune function and for comparative analysis. The results showed that PtCTL-9 displayed broad non-self immunity. PtCTL-9 could also function as an opsonin to promote phagocytosis and the in vitro encapsulation of hemocytes. These results indicated that PtCTL-9 has an extensive nonself-recognition ability, regulates pathogen clearance, and its QPN motif is important in PtCTL-9's immune function.


Assuntos
Proteínas de Artrópodes/metabolismo , Braquiúros/imunologia , Lectinas Tipo C/metabolismo , Proteínas Opsonizantes/metabolismo , Motivos de Aminoácidos/genética , Motivos de Aminoácidos/imunologia , Animais , Proteínas de Artrópodes/genética , Braquiúros/genética , Braquiúros/metabolismo , Hemócitos/metabolismo , Imunidade Inata , Lectinas Tipo C/genética , Proteínas Opsonizantes/genética , Fagocitose , Filogenia , Alinhamento de Sequência , Transdução de Sinais/imunologia
14.
ACS Appl Bio Mater ; 4(1): 795-806, 2021 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-33490885

RESUMO

Nanoparticles find increasing applications in life science and biomedicine. The fate of nanoparticles in a biological system is determined by their protein corona, as remodeling of their surface properties through protein adsorption triggers specific recognition such as cell uptake and immune system clearance and nonspecific processes such as aggregation and precipitation. The corona is a result of nanoparticle-protein and protein-protein interactions and is influenced by particle design. The state-of-the-art design of biomedical nanoparticles is the core-shell structure exemplified by superparamagnetic iron oxide nanoparticles (SPIONs) grafted with dense, well-hydrated polymer shells used for biomedical magnetic imaging and therapy. Densely grafted polymer chains form a polymer brush, yielding a highly repulsive barrier to the formation of a protein corona via nonspecific particle-protein interactions. However, recent studies showed that the abundant blood serum protein albumin interacts with dense polymer brush-grafted SPIONs. Herein, we use isothermal titration calorimetry to characterize the nonspecific interactions between human serum albumin, human serum immunoglobulin G, human transferrin, and hen egg lysozyme with monodisperse poly(2-alkyl-2-oxazoline)-grafted SPIONs with different grafting densities and core sizes. These particles show similar protein interactions despite their different "stealth" capabilities in cell culture. The SPIONs resist attractive interactions with lysozymes and transferrins, but they both show a significant exothermic enthalpic and low exothermic entropic interaction with low stoichiometry for albumin and immunoglobulin G. Our results highlight that protein size, flexibility, and charge are important to predict protein corona formation on polymer brush-stabilized nanoparticles.


Assuntos
Nanopartículas de Magnetita/química , Proteínas Opsonizantes/química , Polímeros/química , Albumina Sérica/química , Adsorção , Compostos Férricos/química , Humanos , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Proteínas Opsonizantes/metabolismo , Poliaminas/química , Coroa de Proteína/química , Albumina Sérica/metabolismo , Termodinâmica
15.
Adv Mater ; 33(3): e2006160, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33296121

RESUMO

Nanomedicines that target specific blood cells represent an emerging strategy to improve drug biodistribution. However, the protein corona usually disrupts nanomedicine targeting to cells and tissues. Herein, instead of exploring synthetic methods to mitigate the impact of the protein corona, its natural interactions with blood cells are leveraged and turn the protein corona into an active ingredient in treating lung inflammation. It is discovered that molecularly engineered liposomes with inverse phosphocholine lipids rapidly enrich complement fragment iC3b by "voluntary opsonization," which triggers neutrophil hijacking through complement receptor 3 phagocytosis. This neutrophil targeting is cell-state dependent as only those activated by acute inflammation display efficient neutrophil reconstruction. The liposome-loaded neutrophils migrate across the alveolar-capillary barrier, accumulate in the inflamed lung parenchyma within hours, and release their payloads to kill the bacteria. This work shows that, in addition to biological cells, the protein corona can be a new platform for active and precision nanomedicine.


Assuntos
Nanomedicina/métodos , Proteínas Opsonizantes/metabolismo , Medicina de Precisão/métodos , Engenharia , Inflamação/imunologia , Inflamação/terapia , Neutrófilos/imunologia , Receptores de Complemento/metabolismo
16.
Biochem Biophys Res Commun ; 534: 193-198, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33280820

RESUMO

Vaccination evoking immunity in susceptible individuals has become the most effective solution to combat infectious diseases. The surface antigen of hepatitis B virus (HBsAg) is a mandatory vaccine for children in China. Herein, we designed an antimicrobial protein consisting of an antimicrobial peptide Thanatin at the N-terminus fused with the HBsAg at the C-terminus. The expressed Thanatin-GFP-HBsAg (TGH) quantitively bound with the anti-HBsAg antibody by ELISA, and after exposure to TGH, Gram-negative E. coli cells became fluorescencent indicating the binding of TGH with the bacterial cells. We also demonstrated that TGH could intercalated into the lipid bilayer as shown by the quartz crystal microbalance with dissipation (QCM-D) and TEM. Moreover, the TGH bound E. coli cells attracted anti-HBsAg IgG as shown by the experiments that in turn treated the E. coli cells with TGF, anti-HBsAg serum and PE labelled goat anti-mouse IgG antibodies. After supplementation with serum from HBsAg vaccinated individuals, TGH showed improved bactericidal effect in vitro. In vivo experiments showed that the mice receiving TGH vaccination show quicker clearance of MDR E. coli pretreated with TGH and better survive in comparison with groups treated with piperacillin plus subatan. In addition, anti-HBsAg serum supplementation also improved the endocytosis of TGH decorated bacteria by leukocytes. This study reported a novel solution to combat infectious pathogens based on the membrane penetrating effect of antimicrobial peptides.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Ativação do Complemento , Farmacorresistência Bacteriana Múltipla , Antígenos de Superfície da Hepatite B/imunologia , Fagocitose , Animais , Antibacterianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Membrana Celular/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Antígenos de Superfície da Hepatite B/genética , Antígenos de Superfície da Hepatite B/metabolismo , Vacinas contra Hepatite B , Humanos , Masculino , Camundongos Endogâmicos BALB C , Proteínas Opsonizantes/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo
17.
Elife ; 92020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32876566

RESUMO

HIV transmission via genital and colorectal mucosa are the most common routes of dissemination. Here, we explored the effects of free and complement-opsonized HIV on colorectal tissue. Initially, there was higher antiviral responses in the free HIV compared to complement-opsonized virus. The mucosal transcriptional response at 24 hr revealed the involvement of activated T cells, which was mirrored in cellular responses observed at 96 hr in isolated mucosal T cells. Further, HIV exposure led to skewing of T cell phenotypes predominantly to inflammatory CD4+ T cells, that is Th17 and Th1Th17 subsets. Of note, HIV exposure created an environment that altered the CD8+ T cell phenotype, for example expression of regulatory factors, especially when the virions were opsonized with complement factors. Our findings suggest that HIV-opsonization alters the activation and signaling pathways in the colorectal mucosa, which promotes viral establishment by creating an environment that stimulates mucosal T cell activation and inflammatory Th cells.


Assuntos
Ativação do Complemento/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Mucosa Intestinal/imunologia , Ativação Linfocitária/imunologia , Adolescente , Adulto , Colo/imunologia , Colo/virologia , Proteínas do Sistema Complemento/química , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Feminino , Humanos , Mucosa Intestinal/virologia , Masculino , Proteínas Opsonizantes/química , Proteínas Opsonizantes/imunologia , Proteínas Opsonizantes/metabolismo , Linfócitos T/imunologia , Adulto Jovem
18.
Open Biol ; 10(9): 200192, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32873151

RESUMO

Trichomoniasis is the third most common sexually transmitted infection in humans and is caused by the protozoan parasite, Trichomonas vaginalis (Tv). Pathogenic outcomes are more common in women and generally include mild vaginitis or cervicitis. However, more serious effects associated with trichomoniasis include adverse reproductive outcomes. Like other infectious agents, pathogenesis from Tv infection is predicted to be the result of both parasite and host factors. At the site of infection, neutrophils are the most abundant immune cells present and probably play key roles in both parasite clearance and inflammatory pathology. Here, we discuss the evidence that neutrophils home to the site of Tv infection, kill the parasite, and that in some circumstances, parasites possibly evade neutrophil-directed killing. In vitro, the parasite is killed by neutrophils using a novel antimicrobial mechanism called trogocytosis, which probably involves both innate and adaptive immunity. While mechanisms of evasion are mostly conjecture at present, the persistence of Tv infections in patients argues strongly for their existence. Additionally, many strains of Tv harbour microbial symbionts Mycoplasma hominis or Trichomonasvirus, which are both predicted to impact neutrophil responses against the parasite. Novel research tools, especially animal models, will help to reveal the true outcomes of many factors involved in neutrophil-Tv interactions during trichomoniasis.


Assuntos
Interações Hospedeiro-Parasita/imunologia , Neutrófilos/imunologia , Vaginite por Trichomonas/imunologia , Vaginite por Trichomonas/parasitologia , Trichomonas vaginalis/imunologia , Animais , Apoptose , Quimiotaxia de Leucócito/imunologia , Citotoxicidade Imunológica , Suscetibilidade a Doenças/imunologia , Feminino , Humanos , Evasão da Resposta Imune , Neutrófilos/metabolismo , Neutrófilos/patologia , Proteínas Opsonizantes/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Simbiose , Vaginite por Trichomonas/metabolismo
19.
Mol Immunol ; 127: 67-77, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32927166

RESUMO

Galectins are the family of carbohydrate-binding proteins that participate in host-pathogen interaction. In this study, a galectin-4 homolog (OnGal-4) from Nile tilapia (Oreochromis niloticus) was characterized. The open reading frame of OnGal-4 was 1194 bp, encoding a peptide of 397 amino including two CRD regions and two carbohydrate recognition sites. OnGal-4 mRNA was expressed in all examined tissues with the highest level in spleen. After Streptococcus agalactiae (S.agalactiae) challenge, the OnGal-4 expression was up-regulated in the spleen, head kidney, brain, and monocytes/macrophages (Mo/MΦ). The in vitro experiments showed that recombinant OnGal-4 (rOnGal-4) protein could bind and agglutinate S.agalactiae and A.hydrophila. Also, rOnGal-4 could induce cytokines expressions and increased bactericidal activity of Mo/MΦ. Further in vivo analysis indicated that OnGal-4 overexpression could protect O.niloticus from S.agalactiae infection through modulating inflammation response. Our study suggested that OnGal-4 could improve immune response against bacterial infection by mediating pathogen recognition and opsonization.


Assuntos
Infecções Bacterianas/imunologia , Ciclídeos/imunologia , Ciclídeos/microbiologia , Galectina 4/química , Galectina 4/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade , Proteínas Opsonizantes/metabolismo , Aglutinação , Sequência de Aminoácidos , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/microbiologia , Ciclídeos/genética , Citocinas/metabolismo , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Especificidade de Órgãos , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Sequências Repetitivas de Aminoácidos , Análise de Sobrevida
20.
J Vis Exp ; (162)2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32831311

RESUMO

The protocol describes how to set up and run a flow cytometry-based phagocytosis assay of Plasmodium falciparum-infected erythrocytes (IEs) opsonized by naturally acquired IgG antibodies specific for VAR2CSA. VAR2CSA is the parasite antigen that mediates the selective sequestration of IEs in the placenta that can cause a severe form of malaria in pregnant women, called placental malaria (PM). Protection from PM is mediated by VAR2CSA-specific antibodies that are believed to function by inhibiting placental sequestration and/or by opsonizing IEs for phagocytosis. The assay employs late-stage-synchronized IEs that have been selected in vitro to express VAR2CSA, plasma/serum-antibodies from women with naturally acquired PM-specific immunity, and the phagocytic cell line THP-1. However, the protocol can easily be modified to assay the functionality of antibodies to any parasite antigen present on the IE surface, whether induced by natural exposure or by vaccination. The assay offers simple and high-throughput evaluation, with good reproducibility, of an important functional aspect of antibody-mediated immunity in malaria. It is, therefore, useful when evaluating clinical immunity to P. falciparum malaria, a major cause of morbidity and mortality in the tropics, particularly in sub-Saharan Africa.


Assuntos
Anticorpos Antiprotozoários/análise , Bioensaio/métodos , Citometria de Fluxo/métodos , Parasitos/imunologia , Fagocitose , Plasmodium falciparum/imunologia , Animais , Antígenos de Protozoários/imunologia , Eritrócitos/parasitologia , Feminino , Humanos , Malária Falciparum/sangue , Malária Falciparum/parasitologia , Proteínas Opsonizantes/metabolismo , Gravidez , Receptores Fc/metabolismo , Reprodutibilidade dos Testes , Células THP-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...