Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Anal Chem ; 96(16): 6209-6217, 2024 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-38607319

RESUMEN

Vaccine-induced immune thrombotic thrombocytopenia (VITT) is a rare but dangerous side effect of adenoviral-vectored COVID-19 vaccines. VITT had been linked to production of autoantibodies recognizing platelet factor 4 (PF4). Here, we characterize anti-PF4 antibodies obtained from a VITT patient's blood. Intact mass measurements indicate that a significant fraction of these antibodies represent a limited number of clones. MS analysis of large antibody fragments (the light chain and the Fc/2 and Fd fragments of the heavy chain) confirms the monoclonal nature of this component of the anti-PF4 antibodies repertoire and reveals the presence of a mature complex biantennary N-glycan within the Fd segment. Peptide mapping using two complementary proteases and LC-MS/MS was used to determine the amino acid sequence of the entire light chain and over 98% of the heavy chain (excluding a short N-terminal segment). The sequence analysis allows the monoclonal antibody to be assigned to the IgG2 subclass and verifies that the light chain belongs to the λ-type. Incorporation of enzymatic de-N-glycosylation into the peptide mapping routine allows the N-glycan in the Fab region of the antibody to be localized to the framework 3 region of the VH domain. This novel N-glycosylation site is the result of a single mutation within the germline sequence. Peptide mapping also provides information on lower-abundance (polyclonal) components of the anti-PF4 antibody ensemble, revealing the presence of all four subclasses (IgG1-IgG4) and both types of the light chain (λ and κ). This case study demonstrates the power of combining the intact, middle-down, and bottom-up MS approaches for meaningful characterization of ultralow quantities of pathogenic antibodies extracted directly from patients' blood.


Asunto(s)
Factor Plaquetario 4 , Humanos , Factor Plaquetario 4/inmunología , Factor Plaquetario 4/química , Vacunas contra la COVID-19/efectos adversos , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/química , Autoanticuerpos/inmunología , Autoanticuerpos/sangre , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/química , Secuencia de Aminoácidos , Púrpura Trombocitopénica Trombótica/inducido químicamente , Púrpura Trombocitopénica Trombótica/inmunología
2.
Curr Opin Hematol ; 30(6): 219-229, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37603711

RESUMEN

PURPOSE OF REVIEW: Platelet factor 4 (PF4, CXCL4), the most abundant α-granule platelet-specific chemokine, forms tetramers with an equatorial ring of high positive charge that bind to a wide range of polyanions, after which it changes conformation to expose antigenic epitopes. Antibodies directed against PF4 not only help to clear infection but can also lead to the development of thrombotic disorders such as heparin-induced thrombocytopenia (HIT) and vaccine-induced thrombocytopenia and thrombosis (VITT). This review will outline the different mechanisms through which PF4 engagement with polyanions combats infection but also contributes to the pathogenesis of inflammatory and thrombotic disease states. RECENT FINDINGS: Recent work has shown that PF4 binding to microbial polyanions may improve outcomes in infection by enhancing leukocyte-bacterial binding, tethering pathogens to neutrophil extracellular traps (NETs), decreasing the thrombotic potential of NET DNA, and modulating viral infectivity. However, PF4 binding to nucleic acids may enhance their recognition by innate immune receptors, leading to autoinflammation. Lastly, while HIT is induced by platelet activating antibodies that bind to PF4/polyanion complexes, VITT, which occurs in a small subset of patients treated with COVID-19 adenovirus vector vaccines, is characterized by prothrombotic antibodies that bind to PF4 alone. SUMMARY: Investigating the complex interplay of PF4 and polyanions may provide insights relevant to the treatment of infectious disease while also improving our understanding of the pathogenesis of thrombotic disorders driven by anti-PF4/polyanion and anti-PF4 antibodies.


Asunto(s)
COVID-19 , Trombocitopenia , Humanos , Heparina/efectos adversos , Factor Plaquetario 4/química , Factor Plaquetario 4/metabolismo , Trombocitopenia/patología , Anticuerpos/efectos adversos
3.
Biochemistry ; 62(3): 722-734, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36626574

RESUMEN

Chemokine CXCL4L1, a homologue of CXCL4, is a more potent antiangiogenic ligand. Its structural property is correlated with the downstream receptor binding. The two chemokines execute their functions by binding the receptors of CXCR3A and CXCR3B. The receptors differ by an extra 51-residue extension in the CXCR3B N-terminus. To understand the binding specificity, a GB1 protein scaffold was used to carry different CXCR3 extracellular elements, and artificial CXCL4 and CXCL4L1 monomers were engineered for the binding assay. We first characterized the molten globule property of CXCL4L1. The structural property causes the CXCL4L1 tetramer to dissociate into monomers in low concentrations, but native CXCL4 adopts a stable tetramer structure in solution. In the titration experiments, the combination of the CXCR3A N-terminus and receptor extracellular loop 2 provided moderate and comparable binding affinities to CXCL4 and CXCL4L1, while sulfation on the CXCR3A N-terminal tyrosine residues provided binding specificity. However, the CXCR3B N-terminal extension did not show significant enhancement in the binding of CXCL4 or CXCL4L1. This result indicates that the tendency to form a chemokine monomer and the binding affinity together contribute the high antiangiogenic activity of CXCL4L1.


Asunto(s)
Quimiocinas , Factor Plaquetario 4 , Factor Plaquetario 4/química , Factor Plaquetario 4/metabolismo , Receptores CXCR3/química
4.
Int J Mol Sci ; 22(16)2021 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-34445266

RESUMEN

The rare but dangerous adverse events evidenced after massive vaccination against SARS-CoV-2 are represented by thrombosis and thrombocytopenia. The patients diagnosed with severe COVID-19 may develop a pro-thrombotic state with a much higher frequency, thus we decided to investigate the role of Spike protein (the only common product of the two conditions) or the anti-Spike antibodies in the etiopathogenesis of thrombosis. A pathogenic Platelet Factor 4 (PF4)-dependent syndrome, unrelated to the use of heparin therapy, has been reported after the administration of vaccines in the patients manifesting acute thrombocytopenia and thrombosis. Thus, we aimed at shedding light on the structural similarities of Spike of SARS-CoV-2 and PF4 on their eventual biochemical interactions and on the role of their specific antibodies. The similarities between PF4 and Spike-RBD proteins were evaluated by a comparison of the structures and by testing the cross-reactivity of their specific antibodies by ELISA assays. We found that the anti-Spike antibodies do not recognize PF4, on the contrary, the anti-PF4 antibodies show some cross-reactivity for Spike-RBD. More interestingly, we report for the first time that the PF4 and Spike-RBD proteins can bind each other. These data suggest that the interaction of the two proteins could be involved in the generation of anti-PF4 antibodies, their binding to Spike-RBD, which could lead to platelets aggregation due also to their high expression of ACE2.


Asunto(s)
Vacunas contra la COVID-19/efectos adversos , COVID-19/inmunología , Factor Plaquetario 4 , Glicoproteína de la Espiga del Coronavirus , Reacciones Cruzadas , Humanos , Factor Plaquetario 4/química , Factor Plaquetario 4/inmunología , Unión Proteica , Conformación Proteica , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Trombocitopenia/patología , Trombosis/patología
5.
J Biol Chem ; 293(18): 6869-6882, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29540475

RESUMEN

Platelet factor 4 (PF4) is one of the most abundant cationic proteins secreted from α-granules of activated platelets. Based on its structure, PF4 was assigned to the CXC family of chemokines and has been shown to have numerous effects on myeloid leukocytes. However, the receptor for PF4 remains unknown. Here, we demonstrate that PF4 induces leukocyte responses through the integrin Mac-1 (αMß2, CD11b/CD18). Human neutrophils, monocytes, U937 monocytic and HEK293 cells expressing Mac-1 strongly adhered to immobilized PF4 in a concentration-dependent manner. The cell adhesion was partially blocked by anti-Mac-1 mAb and inhibition was enhanced when anti-Mac-1 antibodies were combined with glycosaminoglycans, suggesting that cell-surface proteoglycans act cooperatively with Mac-1. PF4 also induced Mac-1-dependent migration of human neutrophils and murine WT, but not Mac-1-deficient macrophages. Coating of Escherichia coli bacteria or latex beads with PF4 enhanced their phagocytosis by macrophages by ∼4-fold, and this process was blocked by different Mac-1 antagonists. Furthermore, PF4 potentiated phagocytosis by WT, but not Mac-1-deficient macrophages. As determined by biolayer interferometry, PF4 directly bound the αMI-domain, the major ligand-binding region of Mac-1, and this interaction was governed by a Kd of 1.3 ± 0.2 µm Using the PF4-derived peptide library, synthetic peptides duplicating the αMI-domain recognition sequences and recombinant mutant PF4 fragments, the binding sites for αMI-domain were identified in the PF4 segments Cys12-Ser26 and Ala57-Ser70 These results identify PF4 as a ligand for the integrin Mac-1 and suggest that many immune-modulating effects previously ascribed to PF4 are mediated through its interaction with Mac-1.


Asunto(s)
Leucocitos/metabolismo , Antígeno de Macrófago-1/metabolismo , Factor Plaquetario 4/metabolismo , Animales , Sitios de Unión , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Escherichia coli/inmunología , Células HEK293 , Humanos , Leucocitos/citología , Leucocitos/inmunología , Antígeno de Macrófago-1/genética , Antígeno de Macrófago-1/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Factor Plaquetario 4/química , Factor Plaquetario 4/fisiología
6.
J Formos Med Assoc ; 118(1 Pt 2): 249-259, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29779926

RESUMEN

BACKGROUND/PURPOSE: Abnormal folliculogenesis is one of the cardinal presentations of polycystic ovarian syndrome (PCOS) and permeability of follicular wall has been proposed to be involved in the normal follicular growth. However, whether or not there is a change in intrafollicular permeability underlies PCOS is unknown. METHODS: This was a tertiary center-based case-control study. From 2014 to 2015, thirteen patients with PCOS who underwent in vitro fertilization-embryo transfer (IVF-ET) were enrolled. Eleven normo-ovulatory patients who underwent IVF-ET due to male factor and/or tubal factor infertility were enrolled as the control group. The influence of ovarian follicular fluid (FF) on endothelial cell permeability was evaluated using a human umbilical vein endothelial cell monolayer permeability assay. The intrafollicular expression profiles of angiogenesis-related proteins were analyzed using a Human Angiogenesis Protein Array Kit. RESULTS: The FF from PCOS patients caused significantly poorer endothelial cell permeability comparing with the effect of FF from the control group (46% ± 12% vs. 58% ± 9%, P = 0.023). Among the 55 angiogenesis-related proteins tested, there was a significantly higher level of intrafollicular platelet factor 4 (PF4) and PF4/IL-8 complex in the PCOS group (p = 0.004). The anti-permeability effect of PF4 was related to the decrease in the intercellular gaps and antagonistic binding with IL-8. CONCLUSION: Our study provides the first evidence of the pathophysiologic contribution of the well-known angiostatic protein, PF4, on human reproductive biology. The increase of the intrafollicular PF4 and its anti-permeability effect might affect the formation of FF and folliculogenesis in PCOS.


Asunto(s)
Líquido Folicular/química , Infertilidad Femenina/patología , Factor Plaquetario 4/química , Síndrome del Ovario Poliquístico/patología , Adulto , Estudios de Casos y Controles , Femenino , Fertilización In Vitro , Humanos , Permeabilidad , Taiwán , Centros de Atención Terciaria
8.
Blood ; 125(11): 1826-9, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25595736

RESUMEN

Antibodies specific for platelet factor 4 (PF4)/heparin complexes are central to the pathogenesis of heparin-induced thrombocytopenia. Marginal zone B cells appear to be the source of such antibodies, but whether T-cell help is required is unclear. Here, we showed that induction of PF4/heparin-specific antibodies by PF4/heparin complexes was markedly impaired in mice depleted of CD4 T cells by anti-CD4 antibodies. Furthermore, Rag1-deficient recipient mice produced PF4/heparin-specific antibodies upon PF4/heparin challenge when reconstituted with a mixture of wild-type splenic B cells and splenocytes from B-cell-deficient (µMT) mice but not splenocytes from T- and B-cell-deficient (Rag1 knockout) mice. Lastly, mice with B cells lacking CD40, a B-cell costimulatory molecule that helps T-cell-dependent B-cell responses, displayed a marked reduction of PF4/heparin-specific antibody production following PF4/heparin challenge. Together, these findings show that helper T cells play a critical role in production of PF4/heparin-specific antibodies.


Asunto(s)
Formación de Anticuerpos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Heparina/inmunología , Factor Plaquetario 4/inmunología , Traslado Adoptivo , Animales , Especificidad de Anticuerpos , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Heparina/efectos adversos , Heparina/química , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Humanos , Inmunización , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor Plaquetario 4/química , Trombocitopenia/sangre , Trombocitopenia/etiología , Trombocitopenia/inmunología , Quimera por Trasplante
9.
Blood ; 125(1): 155-61, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25342714

RESUMEN

Antibodies specific for platelet factor 4 (PF4)/heparin complexes are the hallmark of heparin-induced thrombocytopenia and thrombosis (HIT), but many antibody-positive patients have normal platelet counts. The basis for this is not fully understood, but it is believed that antibodies testing positive in the serotonin release assay (SRA) are the most likely to cause disease. We addressed this issue by characterizing PF4-dependent binding of HIT antibodies to intact platelets and found that most antibodies testing positive in the SRA, but none of those testing negative, bind to and activate platelets when PF4 is present without any requirement for heparin (P < .0001). Binding of SRA-positive antibodies to platelets was inhibited by chondroitinase ABC digestion (P < .05) and by the addition of chondroitin-4-sulfate (CS) or heparin in excess quantities. The findings suggest that although all HIT antibodies recognize PF4 in a complex with heparin, only a subset of these antibodies recognize more subtle epitopes induced in PF4 when it binds to CS, the major platelet glycosaminoglycan. Antibodies having this property could explain "delayed HIT" seen in some individuals after discontinuation of heparin and the high risk for thrombosis that persists for weeks in patients recovered from HIT.


Asunto(s)
Anticuerpos/química , Plaquetas/inmunología , Heparina/química , Factor Plaquetario 4/química , Trombocitopenia/inducido químicamente , Trombocitopenia/inmunología , Sulfatos de Condroitina/química , Ensayo de Inmunoadsorción Enzimática , Epítopos/química , Glicosaminoglicanos/química , Humanos , Inmunoglobulina G/química , Selectina-P/química , Activación Plaquetaria , Polisacárido Liasas/química , Unión Proteica , Serotonina/química , Trombosis/inducido químicamente
10.
Int J Mol Sci ; 18(4)2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28368308

RESUMEN

Chemokines mediate diverse fundamental biological processes, including combating infection. Multiple chemokines are expressed at the site of infection; thus chemokine synergy by heterodimer formation may play a role in determining function. Chemokine function involves interactions with G-protein-coupled receptors and sulfated glycosaminoglycans (GAG). However, very little is known regarding heterodimer structural features and receptor and GAG interactions. Solution nuclear magnetic resonance (NMR) and molecular dynamics characterization of platelet-derived chemokine CXCL7 heterodimerization with chemokines CXCL1, CXCL4, and CXCL8 indicated that packing interactions promote CXCL7-CXCL1 and CXCL7-CXCL4 heterodimers, and electrostatic repulsive interactions disfavor the CXCL7-CXCL8 heterodimer. As characterizing the native heterodimer is challenging due to interference from monomers and homodimers, we engineered a "trapped" disulfide-linked CXCL7-CXCL1 heterodimer. NMR and modeling studies indicated that GAG heparin binding to the heterodimer is distinctly different from the CXCL7 monomer and that the GAG-bound heterodimer is unlikely to bind the receptor. Interestingly, the trapped heterodimer was highly active in a Ca2+ release assay. These data collectively suggest that GAG interactions play a prominent role in determining heterodimer function in vivo. Further, this study provides proof-of-concept that the disulfide trapping strategy can serve as a valuable tool for characterizing the structural and functional features of a chemokine heterodimer.


Asunto(s)
Glicosaminoglicanos/química , Simulación de Dinámica Molecular , Dominios Proteicos , Multimerización de Proteína , beta-Tromboglobulina/química , Secuencia de Aminoácidos , Sitios de Unión/genética , Calcio/metabolismo , Quimiocina CXCL1/química , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Glicosaminoglicanos/metabolismo , Células HL-60 , Heparina/química , Heparina/metabolismo , Humanos , Interleucina-8/química , Interleucina-8/genética , Interleucina-8/metabolismo , Cinética , Espectroscopía de Resonancia Magnética , Oligosacáridos/química , Oligosacáridos/metabolismo , Factor Plaquetario 4/química , Factor Plaquetario 4/genética , Factor Plaquetario 4/metabolismo , Unión Proteica , Homología de Secuencia de Aminoácido , beta-Tromboglobulina/genética , beta-Tromboglobulina/metabolismo
11.
Curr Opin Hematol ; 23(5): 462-70, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27380556

RESUMEN

PURPOSE OF REVIEW: The purpose of this review is to summarize recent findings on heparin-induced thrombocytopenia (HIT), a prothrombotic disorder caused by platelet-activating IgG targeting platelet factor 4 (PF4)/polyanion complexes. RECENT FINDINGS: HIT can explain unusual clinical events, including adrenal hemorrhages, arterial/intracardiac thrombosis, skin necrosis, anaphylactoid reactions, and disseminated intravascular coagulation. Sometimes, HIT begins/worsens after stopping heparin ('delayed-onset' HIT). Various HIT-mimicking disorders are recognized (e.g., acute disseminated intravascular coagulation/'shock liver' with limb ischemia). HIT has features of both B-cell and T-cell immune responses; uptake of PF4/heparin complexes into macrophages ('macropinocytosis') facilitates the anti-PF4/heparin immune response. Antibody-induced activation of monocytes and platelets via their FcγIIA receptors triggers an intense procoagulant response. Sometimes, HIT antibodies recognize PF4 bound to (platelet-associated) chondroitin sulfate, explaining how HIT might occur without concurrent or recent heparin (delayed-onset HIT, 'spontaneous HIT syndrome'). The molecular structure of HIT antigen(s) has been characterized, providing a rationale for future drug design to avoid HIT and improve its treatment. The poor correlation between partial thromboplastin time and plasma argatroban levels (risking subtherapeutic anticoagulation) and need for intravenous administration of argatroban have led to increasing 'off-label' treatment with fondaparinux or one of the direct oral anticoagulants. SUMMARY: Understanding the molecular mechanisms and unusual clinical features of HIT will improve its management.


Asunto(s)
Anticoagulantes/efectos adversos , Heparina/efectos adversos , Trombocitopenia/diagnóstico , Trombocitopenia/etiología , Trombocitopenia/terapia , Animales , Autoinmunidad , Plaquetas/metabolismo , Proteínas Portadoras/metabolismo , Terapia Combinada , Manejo de la Enfermedad , Humanos , Neoplasias/sangre , Neoplasias/complicaciones , Activación Plaquetaria , Factor Plaquetario 4/química , Factor Plaquetario 4/metabolismo , Unión Proteica
13.
Blood ; 124(15): 2442-9, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25150299

RESUMEN

The chemokine platelet factor 4 (PF4) undergoes conformational changes when complexing with polyanions. This can induce the antibody-mediated adverse drug effect of heparin-induced thrombocytopenia (HIT). Understanding why the endogenous protein PF4 becomes immunogenic when complexing with heparin is important for the development of other negatively charged drugs and may also hint toward more general mechanisms underlying the induction of autoantibodies to other proteins. By circular dichroism spectroscopy, atomic force microscopy, and isothermal titration calorimetry we characterized the interaction of PF4 with unfractionated heparin (UFH), its 16-, 8-, and 6-mer subfractions, low-molecular-weight heparin (LMWH), and the pentasaccharide fondaparinux. To bind anti-PF4/heparin antibodies, PF4/heparin complexes require (1) an increase in PF4 antiparallel ß-sheets exceeding ∼30% (achieved by UFH, LMWH, 16-, 8-, 6-mer), (2) formation of multimolecular complexes (UFH, 16-, 8-mer), and (3) energy (needed for a conformational change), which is released by binding of ≥11-mer heparins to PF4, but not by smaller heparins. These findings may help to synthesize safer heparins. Beyond PF4 and HIT, the methods applied in the current study may be relevant to unravel mechanisms making other endogenous proteins more vulnerable to undergo conformational changes with little energy requirement (eg, point mutations and post-translational modifications) and thereby predisposing them to become immunogenic.


Asunto(s)
Anticuerpos/metabolismo , Factor Plaquetario 4/química , Factor Plaquetario 4/metabolismo , Calorimetría , Dicroismo Circular , Ensayo de Inmunoadsorción Enzimática , Fondaparinux , Heparina de Bajo-Peso-Molecular/química , Humanos , Microscopía de Fuerza Atómica , Polisacáridos/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Termodinámica
14.
Rinsho Ketsueki ; 57(10): 2124-2135, 2016.
Artículo en Japonés | MEDLINE | ID: mdl-27795522

RESUMEN

Heparin-induced thrombocytopenia (HIT) is a pro-thrombotic side effect of heparin therapy caused by HIT antibodies with platelet-activating properties. Recent advances in understanding of spontaneous HIT syndrome, which can occur even without any heparin exposure despite its clinical and serological characteristics being similar to those of HIT, reveal the following HIT clinical features atypical for an immune-mediated disease. Heparin-naïve patients can develop IgG antibodies as early as day 4, as in a secondary immune response. Evidence for an anamnestic response upon heparin re-exposure is lacking. In addition, HIT antibodies are relatively short-lived, unlike those in a secondary immune response. Antigen immunoassays are commonly used worldwide for serological diagnosis of HIT. However, such assays do not indicate whether HIT antibodies have platelet-activating properties, leading to low diagnostic specificity for HIT. The detection of platelet-activating antibodies using a washed platelet activation assay is crucial for making a HIT diagnosis. These atypical clinical and serological features should be carefully considered while appropriately diagnosing HIT, which leads to appropriate therapy such as immediate administration of an alternative anticoagulant for preventing thromboembolic events and re-administration of heparin during surgery involving cardiopulmonary bypass when HIT antibodies are no longer detectable.


Asunto(s)
Trombocitopenia , Enfermedad Aguda , Animales , Fondaparinux , Heparina/efectos adversos , Humanos , Factor Plaquetario 4/química , Factor Plaquetario 4/metabolismo , Polisacáridos/química , Polisacáridos/metabolismo , Trombocitopenia/diagnóstico , Trombocitopenia/inmunología , Trombocitopenia/terapia
15.
J Biol Chem ; 288(19): 13522-33, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23536183

RESUMEN

BACKGROUND: CXCL4L1 is a highly potent anti-angiogenic and anti-tumor chemokine, and its structural information is unknown. RESULTS: CXCL4L1 x-ray structure is determined, and it reveals a previously unrecognized chemokine structure adopting a novel C-terminal helix conformation. CONCLUSION: The alternative helix conformation enhances the anti-angiogenic activity of CXCL4L1 by reducing the glycosaminoglycan binding ability. SIGNIFICANCE: Chemokine C-terminal helix orientation is critical in regulating their functions. Chemokines, a subfamily of cytokines, are small, secreted proteins that mediate a variety of biological processes. Various chemokines adopt remarkable conserved tertiary structure comprising an anti-parallel ß-sheet core domain followed by a C-terminal helix that packs onto the ß-sheet. The conserved structural feature has been considered critical for chemokine function, including binding to cell surface receptor. The recently isolated variant, CXCL4L1, is a homologue of CXCL4 chemokine (or platelet factor 4) with potent anti-angiogenic activity and differed only in three amino acid residues of P58L, K66E, and L67H. In this study we show by x-ray structural determination that CXCL4L1 adopts a previously unrecognized structure at its C terminus. The orientation of the C-terminal helix protrudes into the aqueous space to expose the entire helix. The alternative helix orientation modifies the overall chemokine shape and surface properties. The L67H mutation is mainly responsible for the swing-out effect of the helix, whereas mutations of P58L and K66E only act secondarily. This is the first observation that reports an open conformation of the C-terminal helix in a chemokine. This change leads to a decrease of its glycosaminoglycan binding properties and to an enhancement of its anti-angiogenic and anti-tumor effects. This unique structure is recent in evolution and has allowed CXCL4L1 to gain novel functional properties.


Asunto(s)
Factor Plaquetario 4/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas Angiogénicas/química , Cristalografía por Rayos X , Cistina/química , Ditiotreitol/química , Heparina/química , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Factor Plaquetario 4/genética , Factor Plaquetario 4/fisiología , Unión Proteica , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Sustancias Reductoras/química
16.
J Biol Chem ; 288(46): 33060-70, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24097975

RESUMEN

Heparin-induced thrombocytopenia (HIT) is a thrombotic complication of heparin therapy mediated by antibodies to complexes between platelet factor 4 (PF4) and heparin or cellular glycosaminoglycans. However, only a fraction of patients with anti-PF4-heparin antibodies develop HIT, implying that only a subset of these antibodies is pathogenic. The basis for the pathogenic potential of anti-PF4-heparin antibodies remains unclear. To elucidate the intrinsic PF4-binding properties of HIT-like monoclonal antibody (KKO) versus non-pathogenic antibody (RTO) at the single-molecule level, we utilized optical trap-based force spectroscopy to measure the strength and probability of binding of surface-attached antibodies with oligomeric PF4 to simulate interactions on cells. To mimic the effect of heparin in bringing PF4 complexes into proximity, we chemically cross-linked PF4 tetramers using glutaraldehyde. Analysis of the force histograms revealed that KKO-PF4 interactions had ∼10-fold faster on-rates than RTO-PF4, and apparent equilibrium dissociation constants differed ∼10-fold with similar force-free off-rates (k(off) = 0.0031 and 0.0029 s(-1)). Qualitatively similar results were obtained for KKO and RTO interacting with PF4-heparin complexes. In contrast to WT PF4, KKO and RTO showed lower and similar binding probabilities to cross-linked PF4(K50E), which forms few if any oligomers. Thus, formation of stable PF4 polymers results in much stronger interactions with the pathogenic antibody without a significant effect on the binding of the non-pathogenic antibody. These results suggest a fundamental difference in the antigen-binding mechanisms between model pathogenic and non-pathogenic anti-PF4 antibodies that might underlie their distinct pathophysiological behaviors.


Asunto(s)
Anticuerpos Monoclonales/química , Especificidad de Anticuerpos , Anticoagulantes/química , Autoanticuerpos/química , Heparina/química , Factor Plaquetario 4/química , Anticuerpos Monoclonales/inmunología , Anticoagulantes/efectos adversos , Anticoagulantes/inmunología , Anticoagulantes/uso terapéutico , Autoanticuerpos/inmunología , Sitios de Unión de Anticuerpos , Heparina/efectos adversos , Heparina/inmunología , Heparina/uso terapéutico , Humanos , Cinética , Factor Plaquetario 4/inmunología , Trombocitopenia/inducido químicamente , Trombocitopenia/inmunología
17.
Am J Physiol Endocrinol Metab ; 307(8): E695-702, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25159325

RESUMEN

Ocular ischemic microenvironment plays a critical role in the progression of diabetic retinopathy (DR). In this study, we investigated the effect of vitreous and aqueous obtained from proliferative DR patients on the function of CD34⁺ cells derived from healthy humans. Human CD34⁺ cells were incubated with vitreous or aqueous of subjects with PDR. After incubation, cell migration of CD34⁺ was evaluated with CXCL12. Intracellular levels of nitric oxide (NO) were measured with DAF-FM. Tube formation assay was used to evaluate the effect of treated CD34⁺ cells on in vitro angiogenesis. Angiogenic protein array and mass spectrometry (MS) were performed to ascertain the factors secreted by healthy nondiabetic CD34⁺ cells exposed to diabetic vitreous or aqueous. PDR vitreous/aqueous reduced migration of CD34⁺ cells (672.45 ± 42.1/736.75 ± 101.7 AFU; P < 0.01) and attenuated intracellular NO levels (182 ± 1.4/184.5 ± 6.3 AFU, P = 0.002). Pretreatment with PDR vitreous suppressed tube formation of human retinal endothelial cells (64 ± 1.6 vs. 80 ± 2.5). CD34⁺ exposed to PDR vitreous resulted in the increased expression of CXCL4 and serpin F1, whereas CD34⁺ exposed to PDR aqueous showed increased expression of CXCL4, serpin F1, and endothelin-1 (ET-1). MS analysis of CD34⁺ (exposed to PDR vitreous) expressed J56 gene segment, isoform 2 of SPARC-related modular calcium-binding protein 2, isoform 1 of uncharacterized protein c1 orf167, integrin α-M, and 40s ribosomal protein s21. Exposure of healthy nondiabetic CD34⁺ cells to PDR vitreous and aqueous resulted in decreased migration, reduced generation of NO, and altered paracrine secretory function. Our results suggest that the contribution of CD34⁺ cells to the aberrant neovascularization observed in PDR is driven more by the proangiogenic effects of the retinal cells rather than the influence of the vitreous.


Asunto(s)
Humor Acuoso/metabolismo , Retinopatía Diabética/metabolismo , Endotelina-1/metabolismo , Proteínas del Ojo/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Factor Plaquetario 4/metabolismo , Serpinas/metabolismo , Vitreorretinopatía Proliferativa/metabolismo , Cuerpo Vítreo/metabolismo , Adulto , Células Madre Adultas/citología , Células Madre Adultas/inmunología , Células Madre Adultas/metabolismo , Células Madre Adultas/patología , Anciano , Antígenos CD34/metabolismo , Humor Acuoso/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células Cultivadas , Quimiotaxis , Retinopatía Diabética/inmunología , Retinopatía Diabética/patología , Endotelina-1/agonistas , Endotelina-1/química , Proteínas del Ojo/agonistas , Proteínas del Ojo/química , Humanos , Persona de Mediana Edad , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Factores de Crecimiento Nervioso/agonistas , Factores de Crecimiento Nervioso/química , Óxido Nítrico/metabolismo , Mapeo Peptídico , Factor Plaquetario 4/agonistas , Factor Plaquetario 4/química , Retina/inmunología , Retina/metabolismo , Retina/patología , Serpinas/agonistas , Serpinas/química , Vitreorretinopatía Proliferativa/inmunología , Vitreorretinopatía Proliferativa/patología , Cuerpo Vítreo/inmunología
18.
Blood ; 120(20): 4134-42, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22966172

RESUMEN

Prevailing approaches to manage autoimmune thrombotic disorders, such as heparin-induced thrombocytopenia, antiphospholipid syndrome and thrombotic thrombocytopenic purpura, include immunosuppression and systemic anticoagulation, though neither provides optimal outcome for many patients. A different approach is suggested by the concurrence of autoantibodies and their antigenic targets in the absence of clinical disease, such as platelet factor 4 in heparin-induced thrombocytopenia and ß(2)-glycoprotein-I (ß(2)GPI) in antiphospholipid syndrome. The presence of autoantibodies in the absence of disease suggests that conformational changes or other alterations in endogenous protein autoantigens are required for recognition by pathogenic autoantibodies. In thrombotic thrombocytopenic purpura, the clinical impact of ADAMTS13 deficiency caused by autoantibodies likely depends on the balance between residual antigen, that is, enzyme activity, and demand imposed by local genesis of ultralarge multimers of von Willebrand factor. A corollary of these concepts is that disrupting platelet factor 4 and ß(2)GPI conformation (or ultralarge multimer of von Willebrand factor oligomerization or function) might provide a disease-targeted approach to prevent thrombosis without systemic anticoagulation or immunosuppression. Validation of this approach requires a deeper understanding of how seemingly normal host proteins become antigenic or undergo changes that increase antibody avidity, and how they can be altered to retain adaptive functions while shedding epitopes prone to elicit harmful autoimmunity.


Asunto(s)
Síndrome Antifosfolípido/terapia , Autoantígenos/inmunología , Terapia Molecular Dirigida , Púrpura Trombocitopénica Idiopática/terapia , Proteínas ADAM/química , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Proteína ADAMTS13 , Especificidad de Anticuerpos , Antígenos de Plaqueta Humana/química , Antígenos de Plaqueta Humana/inmunología , Síndrome Antifosfolípido/inmunología , Autoanticuerpos/inmunología , Autoantígenos/química , Autoantígenos/efectos de los fármacos , Biopolímeros , Heparina/efectos adversos , Humanos , Infecciones/complicaciones , Modelos Moleculares , Factor Plaquetario 4/química , Factor Plaquetario 4/inmunología , Conformación Proteica , Púrpura Trombocitopénica Idiopática/inducido químicamente , Púrpura Trombocitopénica Idiopática/etiología , Púrpura Trombocitopénica Idiopática/inmunología , Vacunas/efectos adversos , beta 2 Glicoproteína I/química , beta 2 Glicoproteína I/inmunología , Factor de von Willebrand/química , Factor de von Willebrand/inmunología
19.
Soft Matter ; 10(16): 2775-84, 2014 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-24667820

RESUMEN

Immunogenicity (i.e., the ability to initiate immune reactions) is one of the major challenges for the development of new drugs, as it may turn the developed drug therapeutically ineffective or cause severe immune-related effects. Using single molecule force spectroscopy, we study rupture forces between the positively charged, endogenous protein platelet factor 4 (PF4; also known as CXC chemokine ligand 4, CXCL4) and the antithrombotic drug heparin and other negatively charged glycosaminoglycans (GAGs), which are known to form immunogenic PF4/GAG-complexes (e.g., heparin and dextran sulfate) as well as non-immunogenic complexes (e.g., chondroitin sulfate A). Our measurements suggest that the average number of sulfate groups per monosaccharide unit (i.e., the degree of sulfation DS) does not affect the unbinding characteristics of single PF4/GAG-bonds (reaction coordinate x0 = 2.2 ± 0.2 Å, energy barrier ΔG ≈ -1 kBT). However, the average number of GAG bonds formed to a single PF4 molecule increases with increasing DS as indicated by a rising frequency of unbinding events, suggesting a multivalent binding scheme between PF4 and GAGs. Our studies show that at least three GAG bonds have to be formed to each PF4 molecule to induce epitope formation on the PF4/GAG-complex to which PF4/GAG-complex specific antibodies bind. Hence, GAG-based drugs that form less than three bonds per PF4 molecule are unlikely to constitute PF4/drug-complexes that are of immunologic relevance.


Asunto(s)
Glicosaminoglicanos/química , Factor Plaquetario 4/química , Humanos , Microscopía de Fuerza Atómica
20.
Biomolecules ; 14(3)2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38540666

RESUMEN

Pathogenic platelet factor 4 (PF4) antibodies contributed to the abnormal coagulation profiles in COVID-19 and vaccinated patients. However, the mechanism of what triggers the body to produce these antibodies has not yet been clarified. Similar patterns and many comparable features between the COVID-19 virus and heparin-induced thrombocytopenia (HIT) have been reported. Previously, we identified a new mechanism of autoimmunity in HIT in which PF4-antibodies self-clustered PF4 and exposed binding epitopes for other pathogenic PF4/eparin antibodies. Here, we first proved that the SARS-CoV-2 spike protein (SP) also binds to PF4. The binding was evidenced by the increase in mass and optical intensity as observed through quartz crystal microbalance and immunosorbent assay, while the switching of the surface zeta potential caused by protein interactions and binding affinity of PF4-SP were evaluated by dynamic light scattering and isothermal spectral shift analysis. Based on our results, we proposed a mechanism for the generation of PF4 antibodies in COVID-19 patients. We further validated the changes in zeta potential and interaction affinity between PF4 and SP and found that their binding mechanism differs from ACE2-SP binding. Importantly, the PF4/SP complexes facilitate the binding of anti-PF4/Heparin antibodies. Our findings offer a fresh perspective on PF4 engagement with the SARS-CoV-2 SP, illuminating the role of PF4/SP complexes in severe thrombotic events.


Asunto(s)
COVID-19 , Trombocitopenia , Humanos , Anticuerpos Monoclonales Humanizados , Factores Inmunológicos , Factor Plaquetario 4/química , Factor Plaquetario 4/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda