Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Biomolecules ; 13(7)2023 07 14.
Article in English | MEDLINE | ID: mdl-37509158

ABSTRACT

Beta-2 microglobulin (B2M) is an immune system protein that is found on the surface of all nucleated human cells. B2M is naturally shed from cell surfaces into the plasma, followed by renal excretion. In patients with impaired renal function, B2M will accumulate in organs and tissues leading to significantly reduced life expectancy and quality of life. While current hemodialysis methods have been successful in managing electrolyte as well as small and large molecule disturbances arising in chronic renal failure, they have shown only modest success in managing plasma levels of B2M and similar sized proteins, while sparing important proteins such as albumin. We describe a systematic protein design effort aimed at adding the ability to selectively remove specific, undesired waste proteins such as B2M from the plasma of chronic renal failure patients. A novel nanoparticle built using a tetrahedral protein assembly as a scaffold that presents 12 copies of a B2M-binding nanobody is described. The designed nanoparticle binds specifically to B2M through protein-protein interactions with nanomolar binding affinity (~4.2 nM). Notably, binding to the nanoparticle increases the effective size of B2M by over 50-fold, offering a potential selective avenue for separation based on size. We present data to support the potential utility of such a nanoparticle for removing B2M from plasma by either size-based filtration or by polyvalent binding to a stationary matrix under blood flow conditions. Such applications could address current shortcomings in the management of problematic mid-sized proteins in chronic renal failure patients.


Subject(s)
Kidney Failure, Chronic , Renal Insufficiency, Chronic , Humans , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/therapy , Quality of Life , Renal Dialysis , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/therapy , beta 2-Microglobulin/metabolism , beta 2-Microglobulin/pharmacology , Nanoparticles/therapeutic use
2.
Cell ; 186(5): 1026-1038.e20, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36868208

ABSTRACT

Down syndrome (DS) is a neurological disorder with multiple immune-related symptoms; however, crosstalk between the CNS and peripheral immune system remains unexplored. Using parabiosis and plasma infusion, we found that blood-borne factors drive synaptic deficits in DS. Proteomic analysis revealed elevation of ß2-microglobulin (B2M), a major histocompatibility complex class I (MHC-I) component, in human DS plasma. Systemic administration of B2M in wild-type mice led to synaptic and memory defects similar to those observed in DS mice. Moreover, genetic ablation of B2m or systemic administration of an anti-B2M antibody counteracts synaptic impairments in DS mice. Mechanistically, we demonstrate that B2M antagonizes NMDA receptor (NMDAR) function through interactions with the GluN1-S2 loop; blocking B2M-NMDAR interactions using competitive peptides restores NMDAR-dependent synaptic function. Our findings identify B2M as an endogenous NMDAR antagonist and reveal a pathophysiological role for circulating B2M in NMDAR dysfunction in DS and related cognitive disorders.


Subject(s)
Down Syndrome , Receptors, N-Methyl-D-Aspartate , beta 2-Microglobulin , Animals , Humans , Mice , beta 2-Microglobulin/metabolism , beta 2-Microglobulin/pharmacology , Cognitive Dysfunction/metabolism , Cross Reactions , Parabiosis , Proteomics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Down Syndrome/blood , Down Syndrome/metabolism
3.
Circ Res ; 128(5): 655-669, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33508948

ABSTRACT

RATIONALE: Circulating monocytes can have proinflammatory or proreparative phenotypes. The endogenous signaling molecules and pathways that regulate monocyte polarization in vivo are poorly understood. We have shown that platelet-derived ß2M (ß-2 microglobulin) and TGF-ß (transforming growth factor ß) have opposing effects on monocytes by inducing inflammatory and reparative phenotypes, respectively, but each bind and signal through the same receptor. We now define the signaling pathways involved. OBJECTIVE: To determine the molecular mechanisms and signal transduction pathways by which ß2M and TGF-ß regulate monocyte responses both in vitro and in vivo. METHODS AND RESULTS: Wild-type- (WT) and platelet-specific ß2M knockout mice were treated intravenously with either ß2M or TGF-ß to increase plasma concentrations to those in cardiovascular diseases. Elevated plasma ß2M increased proinflammatory monocytes, while increased plasma TGFß increased proreparative monocytes. TGF-ßR (TGF-ß receptor) inhibition blunted monocyte responses to both ß2M and TGF-ß in vivo. Using imaging flow cytometry, we found that ß2M decreased monocyte SMAD2/3 nuclear localization, while TGF-ß promoted SMAD nuclear translocation but decreased noncanonical/inflammatory (JNK [jun kinase] and NF-κB [nuclear factor-κB] nuclear localization). This was confirmed in vitro using both imaging flow cytometry and immunoblots. ß2M, but not TGF-ß, promoted ubiquitination of SMAD3 and SMAD4, that inhibited their nuclear trafficking. Inhibition of ubiquitin ligase activity blocked noncanonical SMAD-independent monocyte signaling and skewed monocytes towards a proreparative monocyte response. CONCLUSIONS: Our findings indicate that elevated plasma ß2M and TGF-ß dichotomously polarize monocytes. Furthermore, these immune molecules share a common receptor but induce SMAD-dependent canonical signaling (TGF-ß) versus noncanonical SMAD-independent signaling (ß2M) in a ubiquitin ligase dependent manner. This work has broad implications as ß2M is increased in several inflammatory conditions, while TGF-ß is increased in fibrotic diseases. Graphic Abstract: A graphic abstract is available for this article.


Subject(s)
Monocytes/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , beta 2-Microglobulin/metabolism , Animals , Cell Differentiation , Cells, Cultured , Humans , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/metabolism , Smad Proteins/metabolism , THP-1 Cells , beta 2-Microglobulin/pharmacology
4.
Eur J Pharmacol ; 810: 134-140, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28687195

ABSTRACT

Uremic pruritus is an unpleasant symptom in patients undergoing hemodialysis, and the underlying mechanisms remain unclear. ß2-Microglobulin (ß2-MG) is well-known as an MHC class I molecule and its level is increased in the plasma of patients undergoing hemodialysis. In this study, we investigated whether ß2-MG was a pruritogen in mice. Intradermal injections of ß2-MG into the rostral back induced scratching in a dose-dependent manner. Intradermal injection of ß2-MG into the cheek also elicited scratching, but not wiping. ß2-MG-induced scratching was inhibited by the µ-opioid receptor antagonist naltrexone hydrochloride. ß2-MG-induced scratching was not inhibited by antagonists of itch-related receptors (e.g., H1 histamine receptor (terfenadine), TP thromboxane receptor (DCHCH), BLT1 leukotriene B4 receptor (CMHVA), and proteinase-activated receptor 2 (FSLLRY-NH2)). However, ß2-MG-induced scratching was attenuated in mice desensitized by repeated application of capsaicin and also by a selective transient receptor potential vanilloid 1 (TRPV1) antagonist (BCTC). In addition, ß2-MG induced phosphorylation of extracellular signal-regulated kinase (a marker of activated neurons) in primary culture of dorsal root ganglion neurons that expressed TRPV1. These results suggest that ß2-MG is a pruritogen and elicits itch-related responses, at least in part, through TRPV1-expressing primary sensory neurons.


Subject(s)
Gene Expression Regulation/drug effects , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Pruritus/chemically induced , Pruritus/metabolism , TRPV Cation Channels/metabolism , beta 2-Microglobulin/pharmacology , Animals , Behavior, Animal/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice , Naltrexone/pharmacology , Phosphorylation/drug effects , Pruritus/pathology
5.
Proc Natl Acad Sci U S A ; 113(43): E6630-E6638, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27702900

ABSTRACT

T cells must respond differently to antigens of varying affinity presented at different doses. Previous attempts to map peptide MHC (pMHC) affinity onto T-cell responses have produced inconsistent patterns of responses, preventing formulations of canonical models of T-cell signaling. Here, a systematic analysis of T-cell responses to 1 million-fold variations in both pMHC affinity and dose produced bell-shaped dose-response curves and different optimal pMHC affinities at different pMHC doses. Using sequential model rejection/identification algorithms, we identified a unique, minimal model of cellular signaling incorporating kinetic proofreading with limited signaling coupled to an incoherent feed-forward loop (KPL-IFF) that reproduces these observations. We show that the KPL-IFF model correctly predicts the T-cell response to antigen copresentation. Our work offers a general approach for studying cellular signaling that does not require full details of biochemical pathways.


Subject(s)
HLA-A2 Antigen/immunology , Models, Immunological , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Brefeldin A/pharmacology , Dose-Response Relationship, Immunologic , Gene Expression Regulation , HLA-A2 Antigen/genetics , HLA-A2 Antigen/pharmacology , Humans , Interferon-gamma/pharmacology , Interleukin-2/pharmacology , Jurkat Cells , Kinetics , Lymphocyte Activation/drug effects , Phosphorylation , Primary Cell Culture , Protein Binding , Receptors, Antigen, T-Cell/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology , beta 2-Microglobulin/pharmacology
6.
BMC Nephrol ; 16: 60, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25899529

ABSTRACT

BACKGROUND: The objective of this study was to investigate the influence of ß2-microglobulin (ß2-M) on the epithelial-mesenchymal transition (EMT) in renal tubular epithelial cells. METHODS: A human kidney proximal tubular cell line (HK-2) was used as the proximal tubular cell model. HK-2 cells were exposed to different concentrations of ß2-M (5, 10, 25, and 50 µM) for up to 24, 48 and 72 h. The effects of ß2-M on cell morphology were observed by phase contrast microscopy, and the possible associated mechanisms were assessed by immunofluorescence staining, western blot, RNA interference, immunoprecipitation, and induced coupled plasma mass spectroscopy. RESULTS: ß2-M induced marked morphological alterations in the HK-2 cells, accompanied by the increased expression of extracellular matrix components and α-smooth muscle actin (α-SMA), vimentin and fibronectin and the reduced expression of E-cadherin. Our results also revealed that ß2-M could induce the EMT in the HK-2 cells without significant affecting cell viability. Excess ß2-M in the HK-2 cells led to a decrease in iron and an increase in hypoxia inducible factor-1α (HIF-1α), which induced EMT in the HK-2 cells. Additionally, disrupting the function of the ß2-M/hemochromatosis (HFE) complex by HFE knockdown was sufficient to reverse ß2-M-mediated EMT in the HK-2 cells. CONCLUSION: These findings demonstrate that the activity of ß2-M is mediated by the ß2-M/HFE complex, which regulates intracellular iron homeostasis and HIF-1α and ultimately induces EMT in HK2 cells.


Subject(s)
Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Kidney Tubules, Proximal/drug effects , beta 2-Microglobulin/pharmacology , Actins/drug effects , Actins/metabolism , Blotting, Western , Cadherins/drug effects , Cadherins/metabolism , Cell Line , Epithelial Cells/metabolism , Fibronectins/drug effects , Fibronectins/metabolism , Gene Knockdown Techniques , Hemochromatosis Protein , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoprecipitation , In Vitro Techniques , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mass Spectrometry , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Microscopy, Phase-Contrast , RNA Interference , Vimentin/drug effects , Vimentin/metabolism
7.
PLoS One ; 7(11): e47642, 2012.
Article in English | MEDLINE | ID: mdl-23144825

ABSTRACT

An antibacterial protein (about 12 kDa) was isolated from human amniotic fluid through dialysis, ultrafiltration and C18 reversed-phase HPLC steps. Automated Edman degradation showed that the N-terminal sequence of the antibacterial protein was NH(2)-Ile-Gln-Arg-Thr-Pro-Lys-Ile-Gln-Val-Tyr-Ser-Arg-His-Pro-Ala-Glu-Asn-Gly-. The N-terminal sequence of the antibacterial protein was found to be identical to that of ß(2)-microglobulin, a component of MHC class I molecules, which are present on all nucleated cells. Matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) revealed that the molecular mass of the antibacterial protein was 11,631 Da. This antibacterial protein, ß(2)M, possessed potent antibacterial activity against pathogenic bacteria. Specially, antibacterial activity was observed in potassium buffer, and potassium ion was found to be critical for the antibacterial activity. Interestingly, the antibacterial action of ß(2)M was associated with dissipation of the transmembrane potential, but the protein did not cause damage to the membrane that would result in SYTOX green uptake. In addition, stimulation of WISH amniotic epithelial cells with the bacterial endotoxin lipopolysaccharide (LPS) induced dose-dependent upregulation of ß(2)M mRNA expression. These results suggest that ß(2)M contributes to a self-defense response when amniotic cells are exposed to pathogens.


Subject(s)
Amniotic Fluid/metabolism , Antimicrobial Cationic Peptides/metabolism , beta 2-Microglobulin/metabolism , Amino Acid Sequence , Amniotic Fluid/cytology , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/isolation & purification , Antimicrobial Cationic Peptides/pharmacology , Cells, Cultured , Chromatography, High Pressure Liquid , Drug Resistance, Bacterial , Epithelial Cells/immunology , Epithelial Cells/metabolism , Escherichia coli/drug effects , Escherichia coli/physiology , Female , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Lipopolysaccharides/pharmacology , Listeria monocytogenes/drug effects , Listeria monocytogenes/physiology , Membrane Potentials/drug effects , Microbial Sensitivity Tests , Molecular Sequence Data , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, Protein , beta 2-Microglobulin/genetics , beta 2-Microglobulin/isolation & purification , beta 2-Microglobulin/pharmacology
8.
PLoS One ; 6(11): e27353, 2011.
Article in English | MEDLINE | ID: mdl-22096558

ABSTRACT

The formation of insoluble amyloid fibrils is associated with an array of devastating human diseases. Dialysis-related amyloidosis (DRA) is a severe complication of hemodialysis that results in the progressive destruction of the bones and joints. Elevated concentrations of ß(2)-microglobulin (ß(2)m) in the serum of subjects on hemodialysis promote the formation of amyloid fibrils in the osteoarticular tissues, but the cellular basis for the destruction of these tissues in DRA is poorly understood. In this study we performed a systematic analysis of the interaction of monomeric and fibrillar ß(2)m with primary human cells of the types present in the synovial joints of subjects with DRA. Building upon observations that macrophages infiltrate ß(2)m amyloid deposits in vivo we demonstrate that monocytes, the precursors of macrophages, cannot degrade ß(2)m fibrils, and that both monomeric ß(2)m and fibrillar ß(2)m are cytotoxic to these cells. ß(2)m fibrils also impair the formation of bone resorbing osteoclasts from monocytes and reduce the viability of osteoblasts, the cell type that produces bone. As a consequence, we predict that ß(2)m amyloid will disrupt the remodelling of the bone, which is critical for the maintenance of this tissue. Moreover, we show that ß(2)m fibrils reduce the viability of chondrocytes, rationalizing the loss of cartilage in DRA. Together, our observations demonstrate that ß(2)m cytotoxicity has multiple cellular targets in the osteoarticular tissues and is likely to be a key factor in the bone and joint destruction characteristic of DRA.


Subject(s)
Amyloidosis/metabolism , beta 2-Microglobulin/pharmacology , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Humans , Immunoblotting , Leukocytes, Mononuclear/cytology , Microscopy, Electron, Transmission , Osteoblasts/cytology , Osteoblasts/drug effects , beta 2-Microglobulin/chemistry
9.
Biotechnol Lett ; 31(9): 1361-5, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19466557

ABSTRACT

Multipotent mesenchymal stem cells (MSCs) hold great promise in regenerative medicine, but one of the biggest challenges facing for their application is the ex vivo expansion to obtain enough undifferentiated cells. Fetal bovine serum (FBS), which can elicit possible contaminations of prion, virus, zoonosis or immunological reaction against xenogenic serum antigens, still remains essential to the culture formulations. There is an urgent need to identify potential factors for the undifferentiated expansion of MSCs to reduce the use of FBS or eventually replace it. A previously recognized housekeeping gene, beta2-microglobulin (beta2M), is demonstrated to act as a novel growth factor to stimulate the undifferentiated ex vivo expansion and preserve the pluripotency of adult MSCs from various sources. The use of beta2M might have promising implications for future clinical application of MSCs.


Subject(s)
Cell Proliferation , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , beta 2-Microglobulin/pharmacology , Cell Culture Techniques
10.
Scand J Immunol ; 69(3): 203-12, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19281532

ABSTRACT

Beta2-microglobulin (beta2m) is the light chain of major histocompatibility complex class I (MHC-I) molecules, and is a prerequisite for the binding of peptides to the heavy chain and their presentation to CD8+ T cells. beta2m can be modified in vivo and in vitro by proteolytic cleavage by complement C1 and subsequent carboxypeptidase B-like activity--processes that lead to the generation of desLys(58) beta2m (dbeta2m). This work aims to study the effect of dbeta2m on peptide binding to MHC-I, the influence of dbeta2m on the binding of beta2m to the MHC-I heavy chain and the biological activity of dbeta2m. Both beta2m and dbeta2m are able to support the generation of MHC-I/peptide complexes at 18 degrees C, but complexes formed in the presence of dbeta2m destabilize at 37 degrees C. Moreover, a 250 times higher concentration of dbeta2m than of beta2m is needed to displace MHC-I associated beta2m from the cell surface. In addition, only beta2m is able to restore MHC-I/peptide complex formation on acid-treated cells whereas dbeta2m appears to bind preferentially to denatured MHC-I heavy chains. In cell cultures, exogenously added dbeta2m, but not beta2m, induces apoptotic cell death in monocytic leukaemic cell lines but spares other kinds of leukaemic cells. Additionally, the presence of dbeta2m, and to a lesser extent beta2m, enhances IFN-gamma-induced NO production by monocytic leukaemic cells. In conclusion, these data show that dbeta2m is not able to support the formation of a stable tri-molecular MHC-I complex at physiological temperature and that dbeta2m exerts other biological functions compared to beta2m when bound to cells.


Subject(s)
Apoptosis/physiology , HLA-A Antigens/metabolism , HLA-A2 Antigen/metabolism , Nitric Oxide/biosynthesis , beta 2-Microglobulin/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Binding, Competitive , Cell Survival/drug effects , Cell Survival/physiology , Flow Cytometry , HLA-A Antigens/immunology , HLA-A2 Antigen/immunology , Humans , K562 Cells , Mice , U937 Cells , beta 2-Microglobulin/immunology , beta 2-Microglobulin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL