Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 1224, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38336934

RESUMO

The peripheral immune system is important in neurodegenerative diseases, both in protecting and inflaming the brain, but the underlying mechanisms remain elusive. Alzheimer's Disease is commonly preceded by a prodromal period. Here, we report the presence of large Aß aggregates in plasma from patients with mild cognitive impairment (n = 38). The aggregates are associated with low level Alzheimer's Disease-like brain pathology as observed by 11C-PiB PET and 18F-FTP PET and lowered CD18-rich monocytes. We characterize complement receptor 4 as a strong binder of amyloids and show Aß aggregates are preferentially phagocytosed and stimulate lysosomal activity through this receptor in stem cell-derived microglia. KIM127 integrin activation in monocytes promotes size selective phagocytosis of Aß. Hydrodynamic calculations suggest Aß aggregates associate with vessel walls of the cortical capillaries. In turn, we hypothesize aggregates may provide an adhesion substrate for recruiting CD18-rich monocytes into the cortex. Our results support a role for complement receptor 4 in regulating amyloid homeostasis.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Disfunção Cognitiva , Humanos , Doença de Alzheimer/patologia , Integrina alfaXbeta2 , Monócitos/patologia
2.
Biomacromolecules ; 24(8): 3666-3679, 2023 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-37507377

RESUMO

Survivin, a well-known member of the inhibitor of apoptosis protein family, is upregulated in many cancer cells, which is associated with resistance to chemotherapy. To circumvent this, inhibitors are currently being developed to interfere with the nuclear export of survivin by targeting its protein-protein interaction (PPI) with the export receptor CRM1. Here, we combine for the first time a supramolecular tweezer motif, sequence-defined macromolecular scaffolds, and ultrasmall Au nanoparticles (us-AuNPs) to tailor a high avidity inhibitor targeting the survivin-CRM1 interaction. A series of biophysical and biochemical experiments, including surface plasmon resonance measurements and their multivalent evaluation by EVILFIT, reveal that for divalent macromolecular constructs with increasing linker distance, the longest linkers show superior affinity, slower dissociation, as well as more efficient PPI inhibition. As a drawback, these macromolecular tweezer conjugates do not enter cells, a critical feature for potential applications. The problem is solved by immobilizing the tweezer conjugates onto us-AuNPs, which enables efficient transport into HeLa cells. On the nanoparticles, the tweezer valency rises from 2 to 16 and produces a 100-fold avidity increase. The hierarchical combination of different scaffolds and controlled multivalent presentation of supramolecular binders was the key to the development of highly efficient survivin-CRM1 competitors. This concept may also be useful for other PPIs.


Assuntos
Ouro , Nanopartículas Metálicas , Humanos , Survivina , Células HeLa , Proteínas Inibidoras de Apoptose/metabolismo , Substâncias Macromoleculares/metabolismo , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo
3.
Arthritis Res Ther ; 25(1): 97, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37287025

RESUMO

BACKGROUND: Lymphocyte activation gene-3 (LAG-3) inhibits T cell activation and interferes with the immune response by binding to MHC-II. As antigen presentation is central in rheumatoid arthritis (RA) pathogenesis, we studied aspects of LAG-3 as a serological marker and mediator in the pathogenesis of RA. Since Galectin-3 (Gal-3) is described as an additional binding partner for LAG-3, we also aimed to study the functional importance of this interaction. METHODS: Plasma levels of soluble (s) LAG-3 were measured in early RA patients (eRA, n = 99) at baseline and after 12 months on a treat-to-target protocol, in self-reportedly healthy controls (HC, n = 32), and in paired plasma and synovial fluid (SF) from chronic RA patients (cRA, n = 38). Peripheral blood mononuclear cells (PBMCs) and synovial fluid mononuclear cells (SFMCs) were examined for LAG-3 expression by flow cytometry. The binding and functional outcomes of LAG-3 and Gal-3 interaction were assessed with surface plasmon resonance (SPR) and in cell cultures using rh-LAG3, an antagonistic LAG-3 antibody and a Gal-3 inhibitor. RESULTS: Baseline sLAG-3 in the plasma was increased in eRA compared to HC and remained significantly elevated throughout 12 months of treatment. A high level of sLAG-3 at baseline was associated with the presence of IgM-RF and anti-CCP as well as radiographic progression. In cRA, sLAG-3 was significantly increased in SF compared with plasma, and LAG-3 was primarily expressed by activated T cells in SFMCs compared to PBMCs. Adding recombinant human LAG-3 to RA cell cultures resulted in decreased cytokine secretion, whereas blocking LAG-3 with an antagonistic antibody resulted in increased cytokine secretion. By SPR, we found a dose-dependent binding between LAG-3 and Gal-3. However, inhibiting Gal-3 in cultures did not further change cytokine production. CONCLUSIONS: sLAG-3 in the plasma and synovial fluid is increased in both early and chronic RA patients, particularly in the inflamed joint. High levels of sLAG-3 are associated with autoantibody seropositivity and radiographic progression in eRA, and LAG-3 plays a biologically active role in cRA by decreasing inflammatory cytokine production. This functional outcome is not affected by Gal-3 interference. Our results suggest that LAG-3 is a faceted regulator of inflammation in early and chronic RA.


Assuntos
Artrite Reumatoide , Leucócitos Mononucleares , Humanos , Artrite Reumatoide/metabolismo , Autoanticorpos , Citocinas/metabolismo , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária , Líquido Sinovial/metabolismo
4.
Nat Commun ; 14(1): 976, 2023 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-36813795

RESUMO

Antigen binding by B cell receptors (BCR) on cognate B cells elicits a response that eventually leads to production of antibodies. However, it is unclear what the distribution of BCRs is on the naïve B cell and how antigen binding triggers the first step in BCR signaling. Using DNA-PAINT super-resolution microscopy, we find that most BCRs are present as monomers, dimers, or loosely associated clusters on resting B cells, with a nearest-neighbor inter-Fab distance of 20-30 nm. We leverage a Holliday junction nanoscaffold to engineer monodisperse model antigens with precision-controlled affinity and valency, and find that the antigen exerts agonistic effects on the BCR as a function of increasing affinity and avidity. Monovalent macromolecular antigens can activate the BCR at high concentrations, whereas micromolecular antigens cannot, demonstrating that antigen binding does not directly drive activation. Based on this, we propose a BCR activation model determined by the antigen footprint.


Assuntos
Antígenos , Receptores de Antígenos de Linfócitos B , Receptores de Antígenos de Linfócitos B/metabolismo , Linfócitos B , Ativação Linfocitária , Transdução de Sinais
5.
Scand J Immunol ; 97(2): e13245, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36537046

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease characterized by joint inflammation and bone erosions. The glycosylated programmed death-1 (PD-1) receptor plays an important role in regulating immune responses and maintaining tolerance. In this study, we focus on two features observed in RA: impaired PD-1 signalling and Galectin-3 (Gal-3) upregulation. We hypothesize that Gal-3 binds PD-1 and PD-1 ligands, potentially contributing to impaired PD-1 signalling. PD-1 and Gal-3 levels in RA synovial fluid (SF) and plasma were evaluated by ELISA. PD-1 and Gal-3 interaction was examined by Surface Plasmon Resonance and ELISA. PD-1, PD-L1 and Gal-3 expression on mononuclear cells from SF and peripheral blood as well as fibroblast-like synoviocytes were examined by flow cytometry. Effects of Gal-3 and PD-L1 on osteoclast formation was evaluated by tartrate-resistant acid phosphatase assay. We show that Gal-3 binds PD-1 and PD-L1. Results demonstrated high expression of PD-1 and Gal-3 on mononuclear cells, especially from SF. Gal-3 inhibited PD-1 signalling when PD-L1 was present. Furthermore, a role of Gal-3 in osteoclast formation was observed in vitro, both directly but also through PD-1:PD-L1 inhibition. Effects of Gal-3 on the PD-1 signalling axis are proposed to be inhibitory, meaning high Gal-3 levels in the complex synovial microenvironment are not desirable in RA. Preventing Gal-3's inhibitory role on PD-1 signalling could, therefore, be a therapeutic target in RA by affecting inflammatory T cell responses and osteoclasts.


Assuntos
Artrite Reumatoide , Galectina 3 , Receptor de Morte Celular Programada 1 , Linfócitos T , Humanos , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Antígeno B7-H1/metabolismo , Galectina 3/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patologia , Receptor de Morte Celular Programada 1/metabolismo , Linfócitos T/metabolismo , Linfócitos T/patologia
6.
Front Immunol ; 13: 915890, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35812455

RESUMO

4-1BB is a T cell costimulatory receptor and a member of the tumor necrosis factor receptor superfamily. Here, we show that Galectin-3 (Gal-3) decreases the cellular response to its ligand (4-1BBL). Gal-3 binds to both soluble 4-1BB (s4-1BB) and membrane-bound 4-1BB (mem4-1BB), without blocking co-binding of 4-1BBL. In plasma, we detected complexes composed of 4-1BB and Gal-3 larger than 100 nm in size; these complexes were reduced in synovial fluid from rheumatoid arthritis. Both activated 4-1BB+ T cells and 4-1BB-transfected HEK293 cells depleted these complexes from plasma, followed by increased expression of 4-1BB and Gal-3 on the cell surface. The increase was accompanied by a 4-fold decrease in TNFα production by the 4-1BBhighGal-3+ T cells, after exposure to 4-1BB/Gal-3 complexes. In RA patients, complexes containing 4-1BB/Gal-3 were dramatically reduced in both plasma and SF compared with healthy plasma. These results support that Gal-3 binds to 4-1BB without blocking the co-binding of 4-1BBL. Instead, Gal-3 leads to formation of large soluble 4-1BB/Gal-3 complexes that attach to mem4-1BB on the cell surfaces, resulting in suppression of 4-1BBL's bioactivity.


Assuntos
Galectina 3 , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral , Ligante 4-1BB/química , Ligante 4-1BB/metabolismo , Galectina 3/química , Células HEK293 , Humanos , Receptores de Antígenos de Linfócitos T , Receptores do Fator de Necrose Tumoral/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
7.
J Immunol Methods ; 505: 113277, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35489403

RESUMO

The mass concentration of specific proteins is often used as a biomarker and play an important part in diagnostics of inflammatory diseases. Monodisperse proteins are robustly measured in immunoassays, but it is considerably more complicated to measure polydisperse oligomeric proteins. The degree of protein oligomerization is critical for functional aspects. For such proteins, information on both the mass concentration as well as the degree of oligomerization is important. Here, a time-resolved immunofluorometric assay (TRIFMA) with sensitivity for protein structure to detect homo-oligomeric and polydisperse proteins is presented. An established TRIFMA for mannan-binding lectin (MBL) was modified by implementing an additional blocking step prior to coating with capture antibodies, leading to a decrease in coating density. Recombinant human MBL was sorted into small, intermediate, and large complexes, using gel permeation chromatography. Small MBL complexes were poorly detectable by TRIFMA with a sparse antibody coating, while larger complexes produced a strong response. From comparison of molecular dimensions, this difference can be related to the size of oligomers. In conclusion, it is possible to design oligomer-size-sensitive immunoassays by regulating the inter-molecular distance of capture antibodies on a scale comparable to the size of the oligomers.


Assuntos
Lectina de Ligação a Manose , Cromatografia em Gel , Humanos , Imunoensaio , Testes Imunológicos
8.
Methods Appl Fluoresc ; 10(2)2022 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-35145049

RESUMO

Quantum dots (QDs) have stood out as nanotools for glycobiology due to their photostability and ability to be combined with lectins. Mannose-binding lectin (MBL) is involved in the innate immune system and plays important roles in the activation of the complement cascade, opsonization, and elimination of apoptotic and microbial cells. Herein, adsorption and covalent coupling strategies were evaluated to conjugate QDs to a recombinant human MBL (rhMBL). The most efficient nanoprobe was selected by evaluating the conjugate ability to labelCandida albicansyeasts by flow cytometry. The QDs-rhMBL conjugate obtained by adsorption at pH 6.0 was the most efficient, labelingca.100% of cells with the highest median fluorescence intensity. The conjugation was also supported by Fourier transform infrared spectroscopy, zeta potential, and size analyses.C. albicanslabeling was calcium-dependent; 12% and <1% of cells were labeled in buffers without calcium and containing EDTA, respectively. The conjugate promoted specific labeling (based on cluster effect) since, after inhibition with mannan, there was a reduction of 80% in cell labeling, which did not occur with methyl-α-D-mannopyranoside monosaccharide. Conjugates maintained colloidal stability, bright fluorescence, and biological activity for at least 8 months. Therefore, QDs-rhMBL conjugates are promising nanotools to elucidate the roles of MBL in biological processes.


Assuntos
Pontos Quânticos , Carboidratos , Citometria de Fluxo , Humanos
9.
Arthritis Res Ther ; 24(1): 43, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35164829

RESUMO

OBJECTIVE: During treatment with immune checkpoint inhibitors (ICI) such as the anti-PD-1 antibody pembrolizumab, half of patients with pre-existing inflammatory arthritis experience disease flares. The underlying immunological mechanisms have not been characterized. Here, we investigate the effect of pembrolizumab on cells involved in inflammation and destruction in the synovial joint and how immunosuppressive treatments affect the pembrolizumab-induced immune reactions. METHODS: We included synovial fluid mononuclear cells (SFMCs, n = 28) and peripheral blood mononuclear cells (PBMCs, n = 6) from patients with rheumatoid arthritis and peripheral spondyloarthritis and PBMCs from healthy controls (n = 6). Fibroblast-like synovial cells (FLSs) were grown from SFMCs. The in vitro effect of pembrolizumab was tested in SFMCs cultured for 48 h, FLS-PBMC co-cultures and in SFMCs cultured for 21 days (inflammatory osteoclastogenesis). Cells and supernatants were analyzed by ELISA, flow cytometry, and pro-inflammatory multiplex assay. Finally, the effect of the disease-modifying anti-rheumatic drugs (DMARDs) adalimumab (TNFα inhibitor), tocilizumab (IL-6R inhibitor), tofacitinib (JAK1/JAK3 inhibitor), and baricitinib (JAK1/JAK2 inhibitor) on pembrolizumab-induced immune reactions was tested. RESULTS: Pembrolizumab significantly increased monocyte chemoattractant protein-1 (MCP-1) production by arthritis SFMCs (P = 0.0031) but not by PBMCs from patients or healthy controls (P = 0.77 and P = 0.43). Pembrolizumab did not alter MMP-3 production in FLS-PBMC co-cultures (P = 0.76) or TRAP secretion in the inflammatory osteoclastogenesis model (P = 0.28). In SFMCs, pembrolizumab further increased the production of TNFα (P = 0.0110), IFNγ (P = 0.0125), IL-12p70 (P = 0.0014), IL-10 (P = 0.0100), IL-13 (P = 0.0044), IL-2 (P = 0.0066), and IL-4 (P = 0.0008) but did not change the production of IL-6 (P = 0.1938) and IL-1 (P = 0.1022). The SFMCs treated with pembrolizumab showed an increased frequency of intermediate monocytes (P = 0.044), and the MCP-1 production increased only within the intermediate monocyte subset (P = 0.028). Lastly, adalimumab, baricitinib, and tofacitinib treatment were able to attenuate the pembrolizumab-induced MCP-1 production (P = 0.0004, P = 0.033, and P = 0.025, respectively), while this was not seen with tocilizumab treatment (P = 0.75). CONCLUSION: Pembrolizumab specifically activated intermediate monocytes and induced the production of several cytokines including TNFα but not IL-6. These findings indicate that flares in patients with pre-existing inflammatory arthritis involve monocyte activation and could be managed with TNFα neutralization.


Assuntos
Artrite Reumatoide , Líquido Sinovial , Células Cultivadas , Humanos , Inibidores de Checkpoint Imunológico , Leucócitos Mononucleares , Monócitos , Fator de Necrose Tumoral alfa/farmacologia
10.
Front Immunol ; 13: 1021370, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36591222

RESUMO

Introduction: Many autoimmune diseases are characterized by germinal center (GC)-derived, affinity-matured, class-switched autoantibodies, and strategies to block GC formation and progression are currently being explored clinically. However, extrafollicular responses can also play a role. The aim of this study was to investigate the contribution of the extrafollicular pathway to autoimmune disease development. Methods: We blocked the GC pathway by knocking out the transcription factor Bcl-6 in GC B cells, leaving the extrafollicular pathway intact. We tested the impact of this intervention in two murine models of systemic lupus erythematosus (SLE): a pharmacological model based on chronic epicutaneous application of the Toll-like receptor (TLR)-7 agonist Resiquimod (R848), and 564Igi autoreactive B cell receptor knock-in mice. The B cell intrinsic effects were further investigated in vitro and in autoreactive mixed bone marrow chimeras. Results: GC block failed to curb autoimmune progression in the R848 model based on anti-dsDNA and plasma cell output, superoligomeric DNA complexes, and immune complex deposition in glomeruli. The 564Igi model confirmed this based on anti-dsDNA and plasma cell output. In vitro, loss of Bcl-6 prevented GC B cell expansion and accelerated plasma cell differentiation. In a competitive scenario in vivo, B cells harboring the genetic GC block contributed disproportionately to the plasma cell output. Discussion: We identified the extrafollicular pathway as a key contributor to autoimmune progression. We propose that therapeutic targeting of low quality and poorly controlled extrafollicular responses could be a desirable strategy to curb autoreactivity, as it would leave intact the more stringently controlled and high-quality GC responses providing durable protection against infection.


Assuntos
Autoimunidade , Lúpus Eritematoso Sistêmico , Camundongos , Animais , Linfócitos B , Centro Germinativo , Plasmócitos
11.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34301873

RESUMO

Nanotechnology enables investigations of single biomacromolecules, but technical challenges have limited the application in liquid biopsies, for example, blood plasma. Nonetheless, tools to characterize single molecular species in such samples represent a significant unmet need with the increasing appreciation of the physiological importance of protein structural changes at nanometer scale. Mannose-binding lectin (MBL) is an oligomeric plasma protein and part of the innate immune system through its ability to activate complement. MBL also serves a role as a scavenger for cellular debris, especially DNA. This may link functions of MBL with several inflammatory diseases in which cell-free DNA now appears to play a role, but mechanistic insight has been lacking. By making nanoparticle tracking analysis possible in human plasma, we now show that superoligomeric structures of MBL form nanoparticles with DNA. These oligomers correlate with disease activity in systemic lupus erythematosus patients. With the direct quantification of the hydrodynamic radius, calculations following the principles of Taylor dispersion in the blood stream connect the size of these complexes to endothelial inflammation, which is among the most important morbidities in lupus. Mechanistic insight from an animal model of lupus supported that DNA-stabilized superoligomers stimulate the formation of germinal center B cells and drive loss of immunological tolerance. The formation involves an inverse relationship between the concentration of MBL superoligomers and antibodies to double-stranded DNA. Our approach implicates the structure of DNA-protein nanoparticulates in the pathobiology of autoimmune diseases.


Assuntos
DNA/química , Lúpus Eritematoso Sistêmico/diagnóstico , Nanopartículas/química , Proteínas/química , Adolescente , Adulto , Animais , Linfócitos B , Biomarcadores , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Lectina de Ligação a Manose , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Adulto Jovem
12.
Nat Commun ; 11(1): 4535, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32913217

RESUMO

The current understanding of the biological identity that nanoparticles may acquire in a given biological milieu is mostly inferred from the hard component of the protein corona (HC). The composition of soft corona (SC) proteins and their biological relevance have remained elusive due to the lack of analytical separation methods. Here, we identify a set of specific corona proteins with weak interactions at silica and polystyrene nanoparticles by using an in situ click-chemistry reaction. We show that these SC proteins are present also in the HC, but are specifically enriched after the capture, suggesting that the main distinction between HC and SC is the differential binding strength of the same proteins. Interestingly, the weakly interacting proteins are revealed as modulators of nanoparticle-cell association mainly through their dynamic nature. We therefore highlight that weak interactions of proteins at nanoparticles should be considered when evaluating nano-bio interfaces.


Assuntos
Nanopartículas/química , Coroa de Proteína/química , Química Click , Reagentes de Ligações Cruzadas/química , Células Endoteliais , Humanos , Poliestirenos/química , Ligação Proteica , Coroa de Proteína/análise , Dióxido de Silício/química , Células THP-1
13.
MAbs ; 12(1): 1810488, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32887536

RESUMO

Monoclonal antibodies are a class of biotherapeutics used for an increasing variety of disorders, including cancer, autoimmune, neurodegenerative, and viral diseases. Besides their antigen specificity, therapeutic use also mandates control of their solution interactions and colloidal properties in order to achieve a stable, efficacious, non-immunogenic, and low viscosity antibody solution at concentrations in the range of 50-150 mg/mL. This requires characterization of their reversible self-association, aggregation, and weak attractive and repulsive interactions governing macromolecular distance distributions in solution. Simultaneous measurement of these properties, however, has been hampered by solution nonideality. Based on a recently introduced sedimentation velocity method for measuring macromolecular size distributions in a mean-field approximation for hydrodynamic interactions, we demonstrate simultaneous measurement of polydispersity and weak and strong solution interactions in a panel of antibodies with concentrations up to 45 mg/mL. By allowing approximately an order of magnitude higher concentrations than previously possible in sedimentation velocity size distribution analysis, this approach can substantially improve efficiency and sensitivity for characterizing polydispersity and interactions of therapeutic antibodies at or close to formulation conditions.


Assuntos
Anticorpos Monoclonais/química , Agregados Proteicos , Hidrodinâmica , Concentração de Íons de Hidrogênio , Ultracentrifugação , Viscosidade
14.
J Immunol ; 204(5): 1345-1361, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31969389

RESUMO

Aggregation of α-synuclein (αSN) is an important histological feature of Parkinson disease. Recent studies showed that the release of misfolded αSN from human and rodent neurons is relevant to the progression and spread of αSN pathology. Little is known, however, about the mechanisms responsible for clearance of extracellular αSN. This study found that human complement receptor (CR) 4 selectively bound fibrillar αSN, but not monomeric species. αSN is an abundant protein in the CNS, which potentially could overwhelm clearance of cytotoxic αSN species. The selectivity of CR4 toward binding fibrillar αSN consequently adds an important αSN receptor function for maintenance of brain homeostasis. Based on the recently solved structures of αSN fibrils and the known ligand preference of CR4, we hypothesize that the parallel monomer stacking in fibrillar αSN creates a known danger-associated molecular pattern of stretches of anionic side chains strongly bound by CR4. Conformational change in the receptor regulated tightly clearance of fibrillar αSN by human monocytes. The induced change coupled concomitantly with phagolysosome formation. Data mining of the brain transcriptome in Parkinson disease patients supported CR4 as an active αSN clearance mechanism in this disease. Our results associate an important part of the innate immune system, namely complement receptors, with the central molecular mechanisms of CNS protein aggregation in neurodegenerative disorders.


Assuntos
Integrina alfaXbeta2 , Macrófagos , Doença de Parkinson , Fagossomos , Agregação Patológica de Proteínas , alfa-Sinucleína , Humanos , Integrina alfaXbeta2/química , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Doença de Parkinson/genética , Doença de Parkinson/imunologia , Doença de Parkinson/patologia , Fagossomos/química , Fagossomos/genética , Fagossomos/imunologia , Fagossomos/patologia , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/imunologia , Agregação Patológica de Proteínas/patologia , Estrutura Quaternária de Proteína , alfa-Sinucleína/química , alfa-Sinucleína/genética , alfa-Sinucleína/imunologia
15.
Anal Biochem ; 586: 113414, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31493371

RESUMO

Three-channel 3D printed centerpieces with two sample sectors next to a joint solvent reference sector were recently described as a strategy to double the throughput of sedimentation velocity analytical ultracentrifugation experiments [Anal. Chem. 91 (2019) 5866-5873]. They are compatible with Rayleigh interference optical detection in commercial analytical ultracentrifuges, but require the rotor angles of data acquisition to be repeatedly adjusted during the experiment to record data from the two sample sectors. Here we present an approach to automate this data acquisition mode through the use of a secondary, general-purpose automation software, and an accompanying data pre-processing software for scan sorting.


Assuntos
Impressão Tridimensional/instrumentação , Ultracentrifugação , Automação , Software
16.
Sci Rep ; 7(1): 15653, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29142299

RESUMO

Classic drug development strategies have failed to meet the urgent clinical needs in treating infections with Gram-negative bacteria. Repurposing drugs can lead to timely availability of new antibiotics, accelerated by existing safety profiles. Glatiramer acetate (GA) is a widely used and safe formulation for treatment of multiple sclerosis. It contains a large diversity of essentially isomeric polypeptides with the cationic and amphiphilic character of many antimicrobial peptides (AMP). Here, we report that GA is antibacterial, targeting Gram-negative organisms with higher activity towards Pseudomonas aeruginosa than the naturally-occurring AMP LL-37 in human plasma. As judged from flow cytometric assays, bacterial killing by GA occurred within minutes. Laboratory strains of Escherichia coli and P. aeruginosa were killed by a process of condensing intracellular contents. Efficient killing by GA was also demonstrated in Acinetobacter baumannii clinical isolates and approximately 50% of clinical isolates of P. aeruginosa from chronic airway infection in CF patients. By contrast, the Gram-positive Staphylococcus aureus cells appeared to be protected from GA by an increased formation of nm-scale particulates. Our data identify GA as an attractive drug repurposing candidate to treat infections with Gram-negative bacteria.


Assuntos
Farmacorresistência Bacteriana/genética , Acetato de Glatiramer/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/patogenicidade , Antibacterianos/farmacologia , Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/patogenicidade , Bactérias Gram-Negativas/patogenicidade , Humanos , Fatores Imunológicos/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Testes de Sensibilidade Microbiana , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/patogenicidade , Infecções Estafilocócicas/microbiologia
17.
Biochim Biophys Acta Biomembr ; 1859(3): 425-437, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28064019

RESUMO

The formulation glatiramer acetate (GA) is widely used in therapy of multiple sclerosis. GA consists of random copolymers of four amino acids, in ratios that produce a predominantly positive charge and an amphipathic character. With the extraordinary complexity of the drug, several pharmacological modes-of-action were suggested, but so far none, which rationalizes the cationicity and amphipathicity as part of the mode-of-action. Here, we report that GA rapidly kills primary human T lymphocytes and, less actively, monocytes. LL-37 is a cleavage product of human cathelicidin with important roles in innate immunity. It shares the positive charge and amphipathic character of GA, and, as shown here, also the ability to kill human leukocyte. The cytotoxicity of both compounds depends on sialic acid in the cell membrane. The killing was associated with the generation of CD45+ debris, derived from cell membrane deformation. Nanoparticle tracking analysis confirmed the formation of such debris, even at low GA concentrations. Electric cell-substrate impedance sensing measurements also recorded stable alterations in T lymphocytes following such treatment. LL-37 forms oligomers through weak hydrophobic contacts, which is critical for the lytic properties. In our study, SAXS showed that GA also forms this type of contacts. Taken together, our study offers new insight on the immunomodulatory mode-of-action of positively charged co-polymers. The comparison of LL-37 and GA highlights a consistent requirement of certain oligomeric and chemical properties to support cytotoxic effects of cationic polymers targeting human leukocytes.


Assuntos
Membrana Celular/metabolismo , Acetato de Glatiramer/química , Ácidos Neuramínicos/metabolismo , Polímeros/química , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Apoptose/efeitos dos fármacos , Membrana Celular/química , Células Cultivadas , Citometria de Fluxo , Humanos , Lipossomos/química , Lipossomos/metabolismo , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Ácidos Neuramínicos/química , Polímeros/metabolismo , Polímeros/farmacologia , Espalhamento a Baixo Ângulo , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Difração de Raios X , Catelicidinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...