Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Blood ; 141(10): 1221-1232, 2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36580664

RESUMEN

Type 2B von Willebrand disease (VWD) is an inherited bleeding disorder in which a subset of point mutations in the von Willebrand factor (VWF) A1 domain and recently identified autoinhibitory module (AIM) cause spontaneous binding to glycoprotein Ibα (GPIbα) on the platelet surface. All reported type 2B VWD mutations share this enhanced binding; however, type 2B VWD manifests as variable bleeding complications and platelet levels in patients, depending on the underlying mutation. Understanding how these mutations localizing to a similar region can result in such disparate patient outcomes is essential for detailing our understanding of VWF regulatory and activation mechanisms. In this study, we produced recombinant glycosylated AIM-A1 fragments bearing type 2B VWD mutations and examined how each mutation affects the A1 domain's thermodynamic stability, conformational dynamics, and biomechanical regulation of the AIM. We found that the A1 domain with mutations associated with severe bleeding occupy a higher affinity state correlating with enhanced flexibility in the secondary GPIbα-binding sites. Conversely, mutation P1266L, associated with normal platelet levels, has similar proportions of high-affinity molecules to wild-type (WT) but shares regions of solvent accessibility with both WT and other type 2B VWD mutations. V1316M exhibited exceptional instability and solvent exposure compared with all variants. Lastly, examination of the mechanical stability of each variant revealed variable AIM unfolding. Together, these studies illustrate that the heterogeneity among type 2B VWD mutations is evident in AIM-A1 fragments.


Asunto(s)
Enfermedad de von Willebrand Tipo 2 , Factor de von Willebrand , Humanos , Sitios de Unión , Plaquetas/metabolismo , Mutación , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Enfermedad de von Willebrand Tipo 2/genética , Factor de von Willebrand/química , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
2.
J Comput Chem ; 44(4): 594-601, 2023 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-36398990

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus causing COVID-19, has continued to mutate and spread worldwide despite global vaccination efforts. In particular, the Omicron variant, first identified in South Africa in late November 2021, has become the dominant strain worldwide. Compared to the original strain identified in Wuhan, Omicron features 50 genetic mutations, with 15 mutations in the receptor-binding domain (RBD) of the spike protein, which binds to the human angiotensin-converting enzyme 2 (ACE2) receptor for viral entry. However, it is not completely understood how these mutations alter the interaction and binding strength between the Omicron RBD and ACE2. In this study, we used a combined steered molecular dynamics (SMD) simulation and experimental microscale thermophoresis (MST) approach to quantify the interaction between Omicron RBD and ACE2. We report that the Omicron brings an enhanced RBD-ACE2 interface through N501Y, Q498R, and T478K mutations; the changes further lead to unique interaction patterns, reminiscing the features of previously dominated variants, Alpha (N501Y) and Delta (L452R and T478K). Among the Q493K and Q493R, we report that Q493R shows stronger binding to ACE2 than Q493K due to increased interactions. Our MST data confirmed that the Omicron mutations in RBD are associated with a five-fold higher binding affinity to ACE2 compared to the RBD of the original strain. In conclusion, our results could help explain the Omicron variant's prevalence in human populations, as higher interaction forces or affinity for ACE2 likely promote greater viral binding and internalization, leading to increased infectivity.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Enzima Convertidora de Angiotensina 2 , Simulación de Dinámica Molecular , Mutación , Unión Proteica , SARS-CoV-2/genética
3.
Am J Physiol Heart Circ Physiol ; 322(1): H87-H93, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34890277

RESUMEN

The 2019 coronavirus disease (COVID-19) is the disease caused by SARS-CoV-2 infection. Although this infection has been shown to affect the respiratory system, a high incidence of thrombotic events has been observed in severe cases of COVID-19 and in a significant portion of COVID-19 nonsurvivors. Although prior literature has reported on both the coagulopathy and hypercoagulability of COVID-19, the specifics of coagulation have not been fully investigated. Observations of microthrombosis in patients with COVID-19 have brought attention to potential inflammatory endothelial injury. Von Willebrand factor (VWF) and its protease, A disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), play an important homeostatic role in responding to endothelial injury. This report provides an overview of the literature investigating the role the VWF/ADAMTS13 axis may have in COVID-19 thrombotic events and suggests potential therapeutic strategies to prevent the progression of coagulopathy in patients with COVID-19.


Asunto(s)
Proteína ADAMTS13/metabolismo , Trastornos de la Coagulación Sanguínea/metabolismo , COVID-19/sangre , Factor de von Willebrand/metabolismo , Coagulación Sanguínea , Trastornos de la Coagulación Sanguínea/etiología , COVID-19/complicaciones , Humanos
4.
Biophys J ; 120(10): 1903-1915, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737157

RESUMEN

The globular-to-unraveled conformation transition of von Willebrand factor (vWF), a large polymeric glycoprotein in human blood plasma, is a crucial step in the process of clotting at sites of vascular injury. However, unraveling of vWF multimers in uninjured vasculature can lead to pathology (i.e., thrombus formation or degradation of vWF proteins by enzyme ADAMTS13, making them nonfunctional). To identify blood flow conditions that might induce pathological unraveling of vWF multimers, here we have computed the globular-to-unraveled transition rate of vWF multimers subjected to varying strain rate elongational flow by employing an enhanced sampling technique, the weighted ensemble method. Weighted ensemble sampling was employed instead of standard brute-force simulations because pathological blood flow conditions can induce undesired vWF unraveling on timescales potentially inaccessible to standard simulation methods. Results here indicate that brief but periodic exposure of vWF to the elongational flow of strain rate greater than or equal to 2500 s-1 represents a source of possible pathology caused by the undesired unraveling of vWF multimers.


Asunto(s)
Trombosis , Factor de von Willebrand , Proteína ADAMTS13 , Coagulación Sanguínea , Humanos
5.
Biophys J ; 120(6): 1011-1019, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33607086

RESUMEN

The current COVID-19 pandemic has led to a devastating impact across the world. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (the virus causing COVID-19) is known to use the receptor-binding domain (RBD) at viral surface spike (S) protein to interact with the angiotensin-converting enzyme 2 (ACE2) receptor expressed on many human cell types. The RBD-ACE2 interaction is a crucial step to mediate the host cell entry of SARS-CoV-2. Recent studies indicate that the ACE2 interaction with the SARS-CoV-2 S protein has a higher affinity than its binding with the structurally identical S protein of SARS-CoV-1, the virus causing the 2002-2004 SARS outbreak. However, the biophysical mechanism behind such binding affinity difference is unclear. This study utilizes combined single-molecule force spectroscopy and steered molecular dynamics (SMD) simulation approaches to quantify the specific interactions between SARS-CoV-2 or SARS-CoV-1 RBD and ACE2. Depending on the loading rates, the unbinding forces between SARS-CoV-2 RBD and ACE2 range from 70 to 105 pN and are 30-40% higher than those of SARS-CoV-1 RBD and ACE2 under similar loading rates. SMD results indicate that SARS-CoV-2 RBD interacts with the N-linked glycan on Asn90 of ACE2. This interaction is mostly absent in the SARS-CoV-1 RBD-ACE2 complex. During the SMD simulations, the extra RBD-N-glycan interaction contributes to a greater force and prolonged interaction lifetime. The observation is confirmed by our experimental force spectroscopy study. After removing N-linked glycans on ACE2, its mechanical binding strength with SARS-CoV-2 RBD decreases to a similar level of the SARS-CoV-1 RBD-ACE2 interaction. Together, the study uncovers the mechanism behind the difference in ACE2 binding between SARS-CoV-2 and SARS-CoV-1 and could help develop new strategies to block SARS-CoV-2 entry.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Fenómenos Biomecánicos , Simulación por Computador , Células HEK293 , Humanos , Modelos Biológicos , Polisacáridos/química , Polisacáridos/metabolismo , Unión Proteica , Dominios Proteicos , Imagen Individual de Molécula
6.
Glycobiology ; 31(5): 593-602, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33021626

RESUMEN

Glypican-1 and its heparan sulfate (HS) chains play important roles in modulating many biological processes including growth factor signaling. Glypican-1 is bound to a membrane surface via a glycosylphosphatidylinositol (GPI)-anchor. In this study, we used all-atom molecular modeling and simulation to explore the structure, dynamics, and interactions of GPI-anchored glypican-1, three HS chains, membranes, and ions. The folded glypican-1 core structure is stable, but has substantial degrees of freedom in terms of movement and orientation with respect to the membrane due to the long unstructured C-terminal region linking the core to the GPI-anchor. With unique structural features depending on the extent of sulfation, high flexibility of HS chains can promote multi-site interactions with surrounding molecules near and above the membrane. This study is a first step toward all-atom molecular modeling and simulation of the glycocalyx, as well as its modulation of interactions between growth factors and their receptors.


Asunto(s)
Membrana Celular/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Glipicanos/metabolismo , Heparitina Sulfato/metabolismo , Termodinámica , Membrana Celular/química , Biología Computacional , Glicosilfosfatidilinositoles/química , Glipicanos/química , Heparitina Sulfato/química , Humanos , Modelos Moleculares , Estructura Molecular
7.
Haematologica ; 106(1): 220-229, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31974202

RESUMEN

During infection neuraminidase desialylates platelets and induces their rapid clearance from circulation. The underlying molecular basis, particularly the role of platelet glycoprotein (GP)Ibα therein, is not clear. Utilizing genetically altered mice we report that the extracellular domain of GPIbα, but neither von Willebrand factor nor ADAM17 (a disintegrin and metalloprotease 17), is required for platelet clearance induced by intravenous injection of neuraminidase. Lectin binding to platelets following neuraminidase injection over time revealed that the extent of desialylation of O-glycans correlates with the decrease of platelet count in mice. Injection of α2,3-neuraminidase reduces platelet counts in wild-type but not in transgenic mice expressing only a chimeric GPIbα that misses most of its extracellular domain. Neuraminidase treatment induces unfolding of the O-glycosylated mechanosensory domain in GPIbα as monitored by single-molecule force spectroscopy, increases the exposure of the ADAM17 shedding cleavage site in the mechanosensory domain on the platelet surface, and induces ligand-independent GPIb-IX signaling in human and murine platelets. These results suggest that desialylation of O-glycans of GPIbα induces unfolding of the mechanosensory domain, subsequent GPIb-IX signaling including amplified desialylation of N-glycans, and eventually rapid platelet clearance. This new molecular mechanism of GPIbα-facilitated clearance could potentially resolve many puzzling and seemingly contradicting observations associated with clearance of desialylated or hyposialylated platelets.


Asunto(s)
Plaquetas , Complejo GPIb-IX de Glicoproteína Plaquetaria , Animales , Ratones , Recuento de Plaquetas , Polisacáridos , Transducción de Señal , Factor de von Willebrand
8.
Blood ; 131(7): 787-796, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29203584

RESUMEN

Immune thrombocytopenia (ITP) is a prevalent autoimmune disease characterized by autoantibody-induced platelet clearance. Some ITP patients are refractory to standard immunosuppressive treatments such as intravenous immunoglobulin (IVIg). These patients often have autoantibodies that target the ligand-binding domain (LBD) of glycoprotein Ibα (GPIbα), a major subunit of the platelet mechanoreceptor complex GPIb-IX. However, the molecular mechanism of this Fc-independent platelet clearance is not clear. Here, we report that many anti-LBD monoclonal antibodies such as 6B4, but not AK2, activated GPIb-IX in a shear-dependent manner and induced IVIg-resistant platelet clearance in mice. Single-molecule optical tweezer measurements of antibodies pulling on full-length GPIb-IX demonstrated that the unbinding force needed to dissociate 6B4 from the LBD far exceeds the force required to unfold the juxtamembrane mechanosensory domain (MSD) in GPIbα, unlike the AK2-LBD unbinding force. Binding of 6B4, not AK2, induced shear-dependent unfolding of the MSD on the platelet, as evidenced by increased exposure of a linear sequence therein. Imaging flow cytometry and aggregometry measurements of platelets and LBD-coated platelet-mimetic beads revealed that 6B4 can sustain crosslinking of platelets under shear, whereas 6B4 Fab and AK2 cannot. These results suggest a novel mechanism by which anti-LBD antibodies can exert a pulling force on GPIb-IX via platelet crosslinking, activating GPIb-IX by unfolding its MSD and inducing Fc-independent platelet clearance.


Asunto(s)
Plaquetas/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/farmacología , Inmunoglobulinas Intravenosas/farmacología , Mecanotransducción Celular/efectos de los fármacos , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/etiología , Animales , Anticuerpos Monoclonales/farmacología , Plaquetas/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/fisiología , Mecanotransducción Celular/inmunología , Ratones , Ratones Transgénicos , Complejo GPIb-IX de Glicoproteína Plaquetaria/inmunología , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Púrpura Trombocitopénica Idiopática/inmunología , Resistencia al Corte/efectos de los fármacos , Resistencia al Corte/fisiología , Transducción de Señal/efectos de los fármacos
9.
Soft Matter ; 16(36): 8372-8379, 2020 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-32812974

RESUMEN

The optical responses of molecules and materials provide a basis for chemical measurement and imaging. The optical diffraction limit in conventional light microscopy is exceeded by mechanically probing optical absorption through the photothermal effect with atomic force microscopy (AFM). However, the spatial resolution of AFM-based photothermal optical microscopy is still limited, and the sample surface is prone to damage from scratching due to tip contact, particularly for measurements on soft matter. In this article, we develop peak force visible (PF-vis) microscopy for the measurement of visible optical absorption of soft matter. The spatial resolution of PF-vis microscopy is demonstrated to be 3 nm on green fluorescent protein-labeled virus-like particles, and the imaging sensitivity may approach a single protein molecule. On organic photovoltaic polymers, the spatial distribution of the optical absorption probed by PF-vis microscopy is found to be dependent on the diffusion ranges of excitons in the donor domain. Through finite element modeling and data analysis, the exciton diffusion range of organic photovoltaics can be directly extracted from PF-vis images, saving the need for complex and delicate sample preparations. PF-vis microscopy will enable high-resolution nano-imaging based on light absorption of fluorophores and chromophores, as well as deciphering the correlation between the spatial distribution of photothermal signals and underlying photophysical parameters at the tens of nanometer scale.


Asunto(s)
Colorantes Fluorescentes , Nanotecnología , Proteínas Fluorescentes Verdes , Microscopía de Fuerza Atómica , Polímeros
10.
Biophys J ; 116(10): 1960-1969, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31030883

RESUMEN

In platelets, the glycoprotein (GP) Ib-IX receptor complex senses blood shear flow and transmits the mechanical signals into platelets. Recently, we have discovered a juxtamembrane mechanosensory domain (MSD) within the GPIbα subunit of GPIb-IX. Mechanical unfolding of the MSD activates GPIb-IX signaling into platelets, leading to their activation and clearance. Using optical tweezer-based single-molecule force measurement, we herein report a systematic biomechanical characterization of the MSD in its native, full-length receptor complex and a recombinant, unglycosylated MSD in isolation. The native MSD unfolds at a resting rate of 9 × 10-3 s-1. Upon exposure to pulling forces, MSD unfolding accelerates exponentially over a force scale of 2.0 pN. Importantly, the unfolded MSD can refold with or without applied forces. The unstressed refolding rate of MSD is ∼17 s-1 and slows exponentially over a force scale of 3.7 pN. Our measurements confirm that the MSD is relatively unstable, with a folding free energy of 7.5 kBT. Because MSD refolding may turn off GPIb-IX's mechanosensory signals, our results provide a mechanism for the requirement of a continuous pulling force of >15 pN to fully activate GPIb-IX.


Asunto(s)
Fenómenos Mecánicos , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Replegamiento Proteico , Fenómenos Biomecánicos , Modelos Moleculares , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Dominios Proteicos , Termodinámica
11.
Biophys J ; 116(11): 2092-2102, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31103230

RESUMEN

We perform single-molecule flow experiments using confocal microscopy and a microfluidic device for shear rates up to 20,000 s-1 and present results for the shear-induced unraveling and elongation of tethered von Willebrand factor (VWF) multimers. Further, we employ companion Brownian dynamics simulations to help explain details of our experimental observations using a parameterized coarse-grained model of VWF. We show that global conformational changes of tethered VWF can be accurately captured using a relatively simple mechanical model. Good agreement is found between experimental results and computational predictions for the threshold shear rate of extension, existence of nonhomogenous fluorescence distributions along unraveled multimer contours, and large variations in extensional response behaviors. Brownian dynamics simulations reveal the strong influence of varying chain length, tethering point location, and number of tethering locations on the underlying unraveling response. Through a complex molecule like VWF that naturally adopts a wide distribution of molecular size and has multiple binding sites within each molecule, this work demonstrates the power of tandem experiment and simulation for understanding flow-induced changes in biomechanical state and global conformation of macromolecules.


Asunto(s)
Resistencia al Corte , Factor de von Willebrand/metabolismo , Fenómenos Biomecánicos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Dispositivos Laboratorio en un Chip , Microscopía Fluorescente , Factor de von Willebrand/química
12.
Appl Environ Microbiol ; 85(17)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31253676

RESUMEN

Cryptosporidium parvum causes potentially life-threatening gastrointestinal disease in humans and may not be effectively removed from drinking water via conventional methods. Prior research has shown that environmental biofilms immobilize oocysts from the water column, but the biophysical mechanisms driving this attraction are still under investigation. This study investigates the affinity of C. parvum oocysts to silanized surfaces. Surfaces were prepared with hydroxyl, amine, and carboxyl moieties. Binding forces between the oocysts and these engineered substrates were analyzed, with and without divalent ions, using atomic force microscopy. Binding forces were measured over several weeks to investigate the influence of age on adhesion. C. parvum oocysts bind most strongly to carboxylic acid functional groups, with rupture forces greater than that required to break noncovalent molecular bonds, regardless of oocyst age. This adhesion is shown to be due to divalent cation bridging mechanisms. In addition, the binding strength increases over a 5-week period as the oocysts age, followed by a decrease in the binding strength, which may be related to structural or biochemical changes in the outer wall-bound glycosylated proteins. This study sheds new light on the biochemical parameters that influence C. parvum oocyst binding to surfaces. Increased understanding of how age and water chemistry influence the binding strength of oocysts may inform future developments in environmental detection and drinking water treatment, such as with the development of oocyst-specific sensors that allow for more frequent tracking of oocysts in the environment.IMPORTANCE The mechanisms by which pathogens bind to surfaces are of interest to a wide variety of scientific communities, as these mechanisms drive infectivity, fate, and transport of the pathogenic organisms. This study begins to reveal the mechanism of direct binding of Cryptosporidium parvum to surfaces containing both carboxylic acid and amine moieties, in an attempt to understand how much of the binding ability is due to long-range electrostatic forces versus other mechanisms (specific or nonspecific) of bonding. In addition to improving the scientific understanding of fate and transport of oocysts, an expanded understanding of the binding mechanisms may aid in the development of new tools and sensors designed to detect and track oocysts in waterways. Furthermore, the methods used to examine binding in this study could be translated to other waterborne pathogens of interest.


Asunto(s)
Adhesión Bacteriana , Calcio/metabolismo , Cryptosporidium parvum/fisiología , Agua/química , Biopelículas , Cinética , Oocistos/fisiología , Purificación del Agua
13.
J Chem Phys ; 151(12): 124905, 2019 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-31575216

RESUMEN

The von Willebrand Factor (vWF) is a large blood glycoprotein that aids in hemostasis. Within each vWF monomer, the A2 domain hosts a cleavage site for enzyme ADAMTS13, which regulates the size of vWF multimers. This cleavage site can only be exposed when an A2 domain unfolds, and the unfolding reaction energy landscape is highly sensitive to the force conditions on the domain. Based on previous optical tweezer experimental results, we advance here a new activated A2 monomer model (AA2MM) for coarse-grained modeling of vWF that accurately represents the force-based probabilistic change between the unfolded/refolded states. A system of springs is employed to mimic the complex mechanical response of vWF monomers subject to pulling forces. AA2MM was validated by comparing monomer scale simulation results to data from prior pulling experiments on vWF monomer fragments. The model was further validated by comparing multimer scale Brownian dynamics simulation results to experiments using microfluidic chamber microscopy to visualize tethered vWF proteins subject to flow. The A2 domain unfolding reaction was studied in bulk flow simulations (pure shear and elongation flow), giving evidence that elongational flow drives the vWF size regulation process in blood. The mechanoreactive, coarse-grained AA2MM accurately describes the complex mechanical coupling between human blood flow conditions and vWF protein reactivity.


Asunto(s)
Modelos Químicos , Factor de von Willebrand/química , Proteína ADAMTS13/sangre , Proteína ADAMTS13/química , Fenómenos Biomecánicos , Simulación por Computador , Humanos , Dominios Proteicos , Desplegamiento Proteico
14.
Biophys J ; 114(8): 1816-1829, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29694861

RESUMEN

Von Willebrand factor (VWF) is a large multimeric protein that aids in blood clotting. Near injury sites, hydrodynamic force from increased blood flow elongates VWF, exposing binding sites for platelets and collagen. To investigate VWF binding to collagen that is exposed on injured arterial surfaces, Brownian dynamics simulations are performed with a coarse-grain molecular model. Accounting for hydrodynamic interactions in the presence of a stationary surface, shear flow conditions are modeled. Binding between beads in coarse-grain VWF and collagen sites on the surface is described via reversible ligand-receptor-type bond formation, which is governed via Bell model kinetics. For conditions in which binding is energetically favored, the model predicts a high probability for binding at low shear conditions; this is counter to experimental observations but in agreement with what prior modeling studies have revealed. To address this discrepancy, an additional binding criterion that depends on the conformation of a submonomer feature in the model local to a given VWF binding site is implemented. The modified model predicts shear-induced binding, in very good agreement with experimental observations; this is true even for conditions in which binding is significantly favored energetically. Biological implications of the model modification are discussed in terms of mechanisms of VWF activity.


Asunto(s)
Colágeno/metabolismo , Modelos Moleculares , Resistencia al Corte , Factor de von Willebrand/metabolismo , Fenómenos Biomecánicos , Probabilidad , Unión Proteica
15.
Adv Exp Med Biol ; 1097: 1-27, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30315537

RESUMEN

The endothelial cells (ECs) forming the inner wall of every blood vessel are constantly exposed to the mechanical forces generated by blood flow. The EC responses to these hemodynamic forces play a critical role in the homeostasis of the circulatory system. A variety of mechanosensors and transducers, locating on the EC surface, intra- and trans-EC membrane, and within the EC cytoskeleton, have thus been identified to ensure proper functions of ECs. Among them, the most recent candidate is the endothelial surface glycocalyx (ESG), which is a matrix-like thin layer covering the luminal surface of the EC. It consists of various proteoglycans, glycosaminoglycans, and plasma proteins and is close to other prominent EC mechanosensors and transducers. This chapter summarizes the ESG composition, thickness, and structure observed by different labeling and visualization techniques and in different types of vessels. It also presents the literature in determining the ESG mechanical properties by atomic force microscopy and optical tweezers. The molecular mechanisms by which the ESG plays the role in EC mechanosensing and transduction are described as well as the ESG remodeling by shear stress, the actin cytoskeleton, the membrane rafts, the angiogenic factors, and the sphingosine-1-phosphate.


Asunto(s)
Células Endoteliales/citología , Glicocálix/fisiología , Mecanotransducción Celular , Citoesqueleto de Actina , Proteínas Sanguíneas , Endotelio Vascular , Glicosaminoglicanos , Humanos , Lisofosfolípidos , Microdominios de Membrana , Proteoglicanos , Esfingosina/análogos & derivados , Estrés Mecánico
16.
Blood ; 125(3): 562-9, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25359992

RESUMEN

How glycoprotein (GP)Ib-IX complex on the platelet surface senses the blood flow through its binding to the plasma protein von Willebrand factor (VWF) and transmits a signal into the platelet remains unclear. Here we show that optical tweezer-controlled pulling of the A1 domain of VWF (VWF-A1) on GPIb-IX captured by its cytoplasmic domain induced unfolding of a hitherto unidentified structural domain before the dissociation of VWF-A1 from GPIb-IX. Additional studies using recombinant proteins and mutant complexes confirmed its existence in GPIb-IX and enabled localization of this quasi-stable mechanosensitive domain of ∼60 residues between the macroglycopeptide region and the transmembrane helix of the GPIbα subunit. These results suggest that VWF-mediated pulling under fluid shear induces unfolding of the mechanosensitive domain in GPIb-IX, which may possibly contribute to platelet mechanosensing and/or shear resistance of VWF-platelet interaction. The identification of the mechanosensitive domain in GPIb-IX has significant implications for the pathogenesis and treatment of related blood diseases.


Asunto(s)
Plaquetas/metabolismo , Membrana Celular/metabolismo , Adhesividad Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Estrés Mecánico , Factor de von Willebrand/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Dicroismo Circular , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factor de von Willebrand/química , Factor de von Willebrand/genética
17.
Am J Physiol Cell Physiol ; 311(6): C846-C853, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27681180

RESUMEN

The endothelial surface glycocalyx (ESG) is a carbohydrate-rich layer found on the vascular endothelium, serving critical functions in the mechanotransduction of blood flow-induced forces. One of the most important protective functions of the ESG is to mediate the production of nitric oxide (NO) in response to blood flow. However, the detailed mechanism underlying ESG's mechanotransduction of the production of NO has not been completely identified. Herein, using the cultured rat brain microvascular endothelial cells (bEnd.3) as a model system, we have implemented a combined atomic force and fluorescence microscopy approach to show that the ESG senses and transduces vertical mechanical stretch to produce NO. This rapid NO production is dependent on the presence of both heparan sulfate (HS) and hyaluronic acid (HA) in ESG, as the removal of HS and/or HA leads to a significant decrease in NO production. Moreover, the production of NO is dependent on the intake of Ca2+ via endothelial transient receptor potential (TRP) channels. Together, our results demonstrate the molecular mechanism of rapid production of NO in response to vertical mechanical stretch.


Asunto(s)
Endotelio Vascular/metabolismo , Endotelio Vascular/fisiología , Glicocálix/metabolismo , Glicocálix/fisiología , Mecanotransducción Celular/fisiología , Óxido Nítrico/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Células Cultivadas , Heparitina Sulfato/metabolismo , Ácido Hialurónico/metabolismo , Ratas , Estrés Mecánico
18.
J Mol Recognit ; 28(6): 385-92, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25707989

RESUMEN

The dynamic interactions between leukocyte integrin receptors and ligands in the vascular endothelium, extracellular matrix, or invading pathogens result in leukocyte adhesion, extravasation, and phagocytosis. This work examined the mechanical strength of the connection between iC3b, a complement component that stimulates phagocytosis, and the ligand-binding domain, the I-domain, of integrin αMß2. Single-molecule force measurements of αM I-domain-iC3b complexes were conducted by atomic force microscope. Strikingly, depending on loading rates, immobilization of the I-domain via its C-terminus resulted in a 1.3-fold to 1.5-fold increase in unbinding force compared with I-domains immobilized via the N-terminus. The force spectra (unbinding force versus loading rate) of the I-domain-iC3b complexes revealed that the enhanced mechanical strength is due to a 2.4-fold increase in the lifetime of the I-domain-iC3b bond. Given the structural and functional similarity of all integrin I-domains, our result supports the existing allosteric regulatory model by which the ligand binding strength of integrin can be increased rapidly when a force is allowed to stretch the C-terminus of the I-domain. This type of mechanism may account for the rapid ligand affinity adjustment during leukocyte migration.


Asunto(s)
Complemento C3b/química , Proteínas Inmovilizadas/química , Antígeno de Macrófago-1/química , Humanos , Ligandos , Unión Proteica , Estructura Terciaria de Proteína
20.
J Phys Chem B ; 127(2): 486-494, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36598427

RESUMEN

Viral infection usually begins with adhesion between the viral particle and viral receptors displayed on the cell membrane. The exterior surface of the cell membrane is typically coated with a brush-like layer of molecules, the glycocalyx, that the viruses need to penetrate. Although there is extensive literature on the biomechanics of virus-cell adhesion, much of it is based on continuum-level models that do not address the question of how virus/cell-membrane adhesion occurs through the glycocalyx. In this work, we present a simulation study of the penetration mechanism. Using a coarse-grained molecular model, we study the force-driven and diffusive penetration of a brush-like glycocalyx by viral particles. For force-driven penetration, we find that viral particles smaller than the spacing of molecules in the brush reach the membrane surface readily. For a given maximum force, viral particles larger than the minimum spacing of brush molecules arrest at some distance from the membrane, governed by the balance of elastic and applied forces. For the diffusive case, we find that weak but multivalent attraction between the glycocalyx molecules and the virus effectively leads to its engulfment by the glycocalyx. Our finding provides potential guidance for developing glycocalyx-targeting drugs and therapies by understanding how virus-cell adhesion works.


Asunto(s)
Glicocálix , Virus , Glicocálix/metabolismo , Adhesivos/metabolismo , Membrana Celular/metabolismo , Adhesión Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA