Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 369
Filtrar
1.
Front Immunol ; 11: 1123, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32636836

RESUMEN

Alternative complement pathway (AP) plays an important role in the development of sepsis, which is life threatening. Deficiency of factor H-related protein 1 (FHR-1), which is a regulator of AP, has been considered as a susceptible factor for atypical hemolytic uremic syndrome (aHUS) and other types of nephropathy when an inducer such as infection exists. However, the underlying mechanism of the disease development is largely unknown. There is no report on CFHR1 gene knockout in any animal infection model and its function in vivo is still unclear. Here, a Cfhr1 knockout mouse was generated for investigating AP in sepsis and sepsis-induced acute kidney injury (AKI). We found that murine FHR-1 homolog (FHR-E) deficiency enhanced lipopolysaccharide (LPS)-induced AP activation both in vitro and in vivo and that Cfhr1 knockout mice exhibited more severe sepsis and AKI in response to LPS challenge. These results indicated that FHR-E deficiency promoted LPS-induced sepsis and AKI through AP over-activation, providing a mouse model for studying AP regulation and sepsis. This study revealed the function of FHR-E in vivo, which may further provide hints to the pathogenesis of FHR-1 deficiency-related diseases by enhancing LPS-induced AP activation.


Asunto(s)
Lesión Renal Aguda/etiología , Proteínas Sanguíneas/deficiencia , Sepsis/etiología , Lesión Renal Aguda/inmunología , Secuencia de Aminoácidos , Animales , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/inmunología , Vía Alternativa del Complemento/genética , Vía Alternativa del Complemento/inmunología , Humanos , Técnicas In Vitro , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Filogenia , Sepsis/inmunología , Homología de Secuencia de Aminoácido
2.
Blood ; 136(6): 715-725, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32384141

RESUMEN

Studies of inherited platelet disorders have provided many insights into platelet development and function. Loss of function of neurobeachin-like 2 (NBEAL2) causes gray platelet syndrome (GPS), where the absence of platelet α-granules indicates NBEAL2 is required for their production by precursor megakaryocytes. The endoplasmic reticulum is a dynamic network that interacts with numerous intracellular vesicles and organelles and plays key roles in their development. The megakaryocyte endoplasmic reticulum is extensive, and in this study we investigated its role in the biogenesis of α-granules by focusing on the membrane-resident trafficking protein SEC22B. Coimmunoprecipitation (co-IP) experiments using tagged proteins expressed in human HEK293 and megakaryocytic immortalized megakaryocyte progenitor (imMKCL) cells established binding of NBEAL2 with SEC22B, and demonstrated that NBEAL2 can simultaneously bind SEC22B and P-selectin. NBEAL2-SEC22B binding was also observed for endogenous proteins in human megakaryocytes using co-IP, and immunofluorescence microscopy detected substantial overlap. SEC22B binding was localized to a region of NBEAL2 spanning amino acids 1798 to 1903, where 2 GPS-associated missense variants have been reported: E1833K and R1839C. NBEAL2 containing either variant did not bind SEC22B coexpressed in HEK293 cells. CRISPR/Cas9-mediated knockout of SEC22B in imMKCL cells resulted in decreased NBEAL2, but not vice versa. Loss of either SEC22B or NBEAL2 expression resulted in failure of α-granule production and reduced granule proteins in imMKCL cells. We conclude that SEC22B is required for α-granule biogenesis in megakaryocytes, and that interactions with SEC22B and P-selectin facilitate the essential role of NBEAL2 in granule development and cargo stability.


Asunto(s)
Proteínas Sanguíneas/fisiología , Gránulos Citoplasmáticos/fisiología , Retículo Endoplásmico/fisiología , Megacariocitos/ultraestructura , Biogénesis de Organelos , Proteínas R-SNARE/fisiología , Sitios de Unión , Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/genética , Células Cultivadas , Técnicas de Inactivación de Genes , Síndrome de Plaquetas Grises/genética , Células HEK293 , Humanos , Inmunoprecipitación , Células Progenitoras de Megacariocitos , Megacariocitos/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Mutación Missense , Selectina-P/fisiología , Mapeo de Interacción de Proteínas , Proteínas Recombinantes/metabolismo
3.
Blood Cells Mol Dis ; 79: 102349, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31357175

RESUMEN

Complement factor H (FH) and FH-related proteins (FHRs), structurally similar proteins are involved in the regulation of complement activation. Homozygous deletion of FHR 1 and 3 proteins (FHR1/3-/-) is known as a risk factor for disorders such as aHUS and SLE, characterised by thrombo-inflammatory complications. Interestingly, FHR1/3-/- genotype also exists as polymorphism in healthy population of various ethnicities around the world including 8-10% Indians. In an effort to understand the functional role of this polymorphism, we describe in this study an elevated surface-bound FH on platelets and monocytes, but not other blood cells in FHR1/3 -/- healthy individuals. The FHR1/3-/- platelets displayed diminish ability to form aggregates in response to agonists in vitro. The FHR1/3-/- monocytes displayed elevated secretion of TNFα, IL1ß, IL6 and IL10 in response to TLR ligands. However, exogenous FH limits platelet aggregates formation as well as cytokine secretion in monocytes. Therefore, observations together suggest a differential regulation of platelets and monocytes by FH-FHR1/3 axis in healthy individuals. While these findings will need more detailed investigation, it is clear that the connection between FH-FHR axis and thrombo-inflammatory complications is likely to be complex in diseases including aHUS and SLE, and provide interesting new directions for future study.


Asunto(s)
Plaquetas/fisiología , Proteínas Sanguíneas/deficiencia , Monocitos/fisiología , Proteínas Sanguíneas/metabolismo , Proteínas Inactivadoras del Complemento C3b/metabolismo , Factor H de Complemento/farmacología , Citocinas/metabolismo , Voluntarios Sanos , Humanos , Agregación Plaquetaria
4.
Front Immunol ; 10: 853, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31118930

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a rare disease characterized by microangiopathic hemolytic anemia, thrombocytopenia and renal failure. It is caused by genetic or acquired defects of the complement alternative pathway. Factor H autoantibodies (anti-FHs) have been reported in 10% of aHUS patients and are associated with the deficiency of factor H-related 1 (FHR1). However, FHR1 deficiency is not enough to cause aHUS, since it is also present in about 5% of Caucasian healthy subjects. In this study we evaluated the prevalence of genetic variants in CFH, CD46, CFI, CFB, C3, and THBD in aHUS patients with anti-FHs, using healthy subjects with FHR1 deficiency, here defined "supercontrols," as a reference group. "Supercontrols" are more informative than general population because they share at least one risk factor (FHR1 deficiency) with aHUS patients. We analyzed anti-FHs in 305 patients and 30 were positive. The large majority were children (median age: 7.7 [IQR, 6.6-9.9] years) and 83% lacked FHR1 (n = 25, cases) due to the homozygous CFHR3-CFHR1 deletion (n = 20), or the compound heterozygous CFHR3-CFHR1 and CFHR1-CFHR4 deletions (n = 4), or the heterozygous CFHR3-CFHR1 deletion combined with a frameshift mutation in CFHR1 that generates a premature stop codon (n = 1). Of the 960 healthy adult subjects 48 had the FHR1 deficiency ("supercontrols"). Rare likely pathogenetic variants in CFH, THBD, and C3 were found in 24% of cases (n = 6) compared to 2.1% of the "supercontrols" (P-value = 0.005). We also found that the CFH H3 and the CD46GGAAC haplotypes are not associated with anti-FHs aHUS, whereas these haplotypes are enriched in aHUS patients without anti-FHs, which highlights the differences in the genetic basis of the two forms of the disease. Finally, we confirm that common infections are environmental factors that contribute to the development of anti-FHs aHUS in genetically predisposed individuals, which fits with the sharp peak of incidence during scholar-age. Further studies are needed to fully elucidate the complex genetic and environmental factors underlying anti-FHs aHUS and to establish whether the combination of anti-FHs with likely pathogenetic variants or other risk factors influences disease outcome and response to therapies.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Autoanticuerpos/inmunología , Proteínas Sanguíneas/deficiencia , Proteínas del Sistema Complemento/genética , Síndrome Hemolítico Urémico Atípico/inmunología , Autoantígenos/inmunología , Proteínas Sanguíneas/genética , Niño , Preescolar , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Masculino
5.
J Thromb Haemost ; 17(1): 149-156, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30451376

RESUMEN

Essentials Protein Z (PZ) catalyzes PZ-dependent proteinase inhibitor (ZPI) inactivation of factor (F)Xa. Gene-deletion of PZ or ZPI improves coagulation in hemophilia (FVIII knockout) mice. A PZ blocking antibody enhances thrombin generation in human hemophilia plasma. Suppression of the PZ/ZPI pathway may ameliorate the phenotype of severe hemophilia. SUMMARY: Background Hemostasis requires a balance between procoagulant and anticoagulant factors. Hemophiliacs bleed because of a procoagulant deficiency. Targeted reduction in the activity of endogenous anticoagulant pathways is currently being investigated as a means of improving hemostasis in hemophilia. Protein Z (PZ) is a cofactor that serves as a catalyst for PZ-dependent protease inhibitor (ZPI) inactivation of activated factor X at phospholipid surfaces. Objectives To evaluate the effects of PZ or ZPI gene deletion in hemophilic mice, and of blocking PZ in human hemophilic plasma. Methods A tail vein rebleeding assay (TVRB) was developed on the basis of the serial disruption of clots forming over a period of 15 min following tail vein laceration in an anesthetized mouse. Wild-type (WT)/FVIII knockout FVIIIKO, PZ knockout PZKO/FVIIIKO and ZPI knockout ZPIKO/FVIIIKO mice were evaluated in this model, and their plasmas were tested in thrombin generation assays. A mAb against PZ was evaluated in human hemophilic plasma thrombin generation assays. Results The numbers of clot disruptions (mean ± standard error of the mean) in the TVRB were: 4.0 ± 0.9 for WT/FVIIIKO mice; 23.8 ± 1.1 for WT/FVIIIKO mice supplemented with 100% FVIII; 15.2 ± 1.1 for PZKO/FVIIIKO mice; and 14.7 ± 1.2 for ZPIKO/FVIIIKO mice. Thrombin generation in PZKO/FVIIIKO and ZPIKO/FVIIIKO mouse plasmas was similar to that in FVIIIKO plasma supplemented with ~ 15% recombinant FVIII. A mAb against PZ added to human hemophilic plasma enhanced thrombin generation to an extent similar to the addition of ~ 15% FVIII. Conclusions Blockade of the PZ/ZPI system may be sufficient to ameliorate the phenotype of severe hemophilia.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Coagulación Sanguínea/efectos de los fármacos , Proteínas Sanguíneas/antagonistas & inhibidores , Coagulantes/farmacología , Factor VIII/metabolismo , Factor Xa/metabolismo , Hemofilia A/tratamiento farmacológico , Serpinas/metabolismo , Animales , Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Modelos Animales de Enfermedad , Factor VIII/genética , Predisposición Genética a la Enfermedad , Hemofilia A/sangre , Hemofilia A/genética , Humanos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Prueba de Estudio Conceptual , Serpinas/deficiencia , Serpinas/genética , Trombina/metabolismo
7.
Turk J Pediatr ; 60(3): 270-276, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30511539

RESUMEN

Gür-Çetinkaya P, Çagdas-Ayvaz DN, Öksüz AB, Ertoy A, Hayran U, Özkan F, Erol M, Tezcan I. Advantage of the subcutaneous immunoglobulin replacement therapy in primary immunodeficient patients with or without secondary protein loss. Turk J Pediatr 2018; 60: 270-276. In recent years subcutaneous immunoglobulin is widely used for primary immunodeficient patients. Subcutaneous administration provides a more stable and higher serum immunoglobulin levels due to continuous and steady transition from lymphatics to the systemic circulation. We aimed to evaluate the changes in serum immunoglobulin levels under subcutaneous immunoglobulin therapy in patients with primary immunodeficiency with or without secondary protein loss. Nine patients with primary immunodeficiency who switched to subcutaneous immunoglobulin were enrolled. Age, gender, diagnosis, reasons of transition to subcutaneous route, reasons of secondary protein loss were recorded. A questionnaire consisting of frequencies and types of infections, side effects observed with intravenous and subcutaneous routes; date and reason of transition to subcutaneous route were asked to all participants. Serum immunoglobulin levels at the 3rd and the 6th months before and after subcutaneous route were recorded. Of the 9 patients (M/F=4/5) the median age was 12 years (6.1-28.7) and 5 of them had protein loss. In total, 444 injections were applied, and all patients experienced local reactions. Infections were more frequent under intravenous than subcutaneous route (p=0.004). We observed an increase in immunoglobulin levels under subcutaneous route (p=0.069 at 3rd; p=0.13 at 6th month). This increase was evident at the 3rd month of transition to subcutaneous route in patients with protein loss (p=0.080). There was an increase in serum immunoglobulin levels under subcutaneous route. However, increase was not statistically significant since the study group was small. This increment was prominent in patients with protein loss. Subcutaneous administration may be a good alternative for primary immunodeficient patients with protein loss who have persistent low serum immunoglobulin levels despite increments in the intravenous immunoglobulin doses.


Asunto(s)
Proteínas Sanguíneas/deficiencia , Inmunización Pasiva/métodos , Inmunoglobulina G/administración & dosificación , Síndromes de Inmunodeficiencia/terapia , Administración Intravenosa , Adolescente , Adulto , Niño , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Inmunización Pasiva/efectos adversos , Inmunoglobulina G/sangre , Síndromes de Inmunodeficiencia/complicaciones , Infecciones/epidemiología , Reacción en el Punto de Inyección/epidemiología , Reacción en el Punto de Inyección/etiología , Inyecciones Subcutáneas , Masculino , Estudios Retrospectivos , Encuestas y Cuestionarios , Adulto Joven
8.
Arterioscler Thromb Vasc Biol ; 38(10): 2435-2447, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354215

RESUMEN

Objective- Human and mouse megakaryocytes lacking NBEAL2 (neurobeachin-like 2) produce platelets where α-granules lack protein cargo. This cargo is mostly megakaryocyte-synthesized, but some proteins, including FGN (fibrinogen), are endocytosed. In this study, we examined the trafficking of both types of cargo within primary megakaryocytes cultured from normal and NBEAL2-null mice, to determine the role of NBEAL2 in α-granule maturation. We also examined the interaction of NBEAL2 with the granule-associated protein P-selectin in human megakaryocytes and platelets. Approach and Results- Fluorescence microscopy was used to compare uptake of labeled FGN by normal and NBEAL2-null mouse megakaryocytes, which was similar in both. NBEAL2-null cells, however, showed decreased FGN retention, and studies with biotinylated protein showed rapid loss rather than increased degradation. Intracellular tracking via fluorescence microscopy revealed that in normal megakaryocytes, endocytosed FGN sequentially associated with compartments expressing RAB5 (Ras-related protein in brain 5), RAB7 (Ras-related protein in brain 7), and P-selectin, where it was retained. A similar initial pattern was observed in NBEAL2-null megakaryocytes, but then FGN passed from the P-selectin compartment to RAB11 (Ras-related protein in brain 11)-associated endosomes before release. Megakaryocyte-synthesized VWF (Von Willebrand factor) was observed to follow the same route out of NBEAL2-null cells. Immunofluorescence microscopy revealed intracellular colocalization of NBEAL2 with P-selectin in human megakaryocytes, proplatelets, and platelets. Native NBEAL2 and P-selectin were coimmunoprecipitated from platelets and megakaryocytes. Conclusions- NBEAL2 is not required for FGN uptake by megakaryocytes. NBEAL2 is required for the retention of both endocytosed and megakaryocyte-synthesized proteins by maturing α-granules, and possibly by platelet-borne granules. This function may involve interaction of NBEAL2 with P-selectin.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Endocitosis , Fibrinógeno/metabolismo , Megacariocitos/metabolismo , Animales , Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/genética , Células Cultivadas , Endosomas/metabolismo , Femenino , Masculino , Ratones Noqueados , Selectina-P/metabolismo , Transporte de Proteínas , Vías Secretoras , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Proteínas de Unión a GTP rab7 , Factor de von Willebrand/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 38(8): 1772-1784, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29930006

RESUMEN

Objective- Nbeal2-/- mice, a model of human gray platelet syndrome, have reduced neutrophil granularity and impaired host defense against systemic Staphylococcus aureus infection. We here aimed to study the role of Nbeal2 deficiency in both leukocytes and platelets during gram-negative pneumonia and sepsis. Approach and Results- We studied the role of Nbeal2 in platelets and leukocytes during murine pneumonia and sepsis by Klebsiella pneumoniae. Apart from platelet α-granule deficiency and reduced neutrophil granularity, also monocyte granularity was reduced in Nbeal2-/- mice, whereas plasma levels of MPO (myeloperoxidase), elastase, NGAL (neutrophil gelatinase-associated lipocalin), and MMP-9 (matrix metalloproteinase 9), and leukocyte CD11b expression were increased. Nbeal2-/- leukocytes showed unaltered in vitro antibacterial response and phagocytosis capacity against Klebsiella, and unchanged reactive nitrogen species and cytokine production. Also during Klebsiella pneumonia and sepsis, Nbeal2-/- mice had similar bacterial growth in lung and distant body sites, with enhanced leukocyte migration to the bronchoalveolar space. Despite similar infection-induced inflammation, organ damage was increased in Nbeal2-/- mice, which was also seen during endotoxemia. Platelet-specific Nbeal2 deficiency did not influence leukocyte functions, indicating that Nbeal2 directly modifies leukocytes. Transfusion of Nbeal2-/- but not of Nbeal2+/+ platelets into thrombocytopenic mice was associated with bleeding in the lung but similar host defense, pointing at a role for platelet α-granules in maintaining vascular integrity but not host defense during Klebsiella pneumosepsis. Conclusions- These data show that Nbeal2 deficiency-resulting in gray platelet syndrome-affects platelets, neutrophils, and monocytes, with intact host defense but increased organ damage during gram-negative pneumosepsis.


Asunto(s)
Plaquetas/metabolismo , Proteínas Sanguíneas/deficiencia , Síndrome de Plaquetas Grises/metabolismo , Infecciones por Klebsiella/metabolismo , Klebsiella pneumoniae/patogenicidad , Insuficiencia Multiorgánica/metabolismo , Neumonía Bacteriana/metabolismo , Sepsis/metabolismo , Animales , Plaquetas/microbiología , Proteínas Sanguíneas/genética , Antígeno CD11b/sangre , Modelos Animales de Enfermedad , Femenino , Síndrome de Plaquetas Grises/sangre , Síndrome de Plaquetas Grises/genética , Interacciones Huésped-Patógeno , Infecciones por Klebsiella/sangre , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/crecimiento & desarrollo , Lipocalina 2/sangre , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/microbiología , Insuficiencia Multiorgánica/sangre , Insuficiencia Multiorgánica/genética , Insuficiencia Multiorgánica/microbiología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Elastasa Pancreática/sangre , Peroxidasa/sangre , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transfusión de Plaquetas , Neumonía Bacteriana/sangre , Neumonía Bacteriana/genética , Neumonía Bacteriana/microbiología , Sepsis/sangre , Sepsis/genética , Sepsis/microbiología
10.
Nat Commun ; 9(1): 2329, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29899553

RESUMEN

The ubiquitin regulatory X domain-containing proteins (UBXNs) are likely involved in diverse biological processes. Their physiological functions, however, remain largely unknown. Here we present physiological evidence that UBXN3B positively regulates stimulator-of-interferon genes (STING) signaling. We employ a tamoxifen-inducible Cre-LoxP approach to generate systemic Ubxn3b knockout in adult mice as the Ubxn3b-null mutation is embryonically lethal. Ubxn3b-/-, like Sting-/- mice, are highly susceptible to lethal herpes simplex virus 1 (HSV-1) and vesicular stomatitis virus (VSV) infection, which is correlated with deficient immune responses when compared to Ubxn3b+/+ littermates. HSV-1 and STING agonist-induced immune responses are also reduced in several mouse and human Ubxn3b-/- primary cells. Mechanistic studies demonstrate that UBXN3B interacts with both STING and its E3 ligase TRIM56, and facilitates STING ubiquitination, dimerization, trafficking, and consequent recruitment and phosphorylation of TBK1. These results provide physiological evidence that links the UBXN family with antiviral immune responses.


Asunto(s)
Proteínas Sanguíneas/inmunología , Proteínas de la Membrana/inmunología , Animales , Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/genética , Células Cultivadas , Femenino , Células HEK293 , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Humanos , Interferón Tipo I/biosíntesis , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Vesiculovirus/inmunología , Vesiculovirus/patogenicidad
11.
Life Sci Space Res (Amst) ; 17: 83-90, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29753417

RESUMEN

Little is known about plasma proteins that can be used as biomarkers for early and late responses to radiation. The purpose of this study was to determine a link between depletion of plasma gelsolin (pGSN) and cell-death as well as inflammatory responses in the lung (one of the tissues known to be radiosensitive) of the same exposed CBA/CaJ mice after exposure to heavy silicon (28Si) ions. To prevent the development of multiple organ dysfunctions, pGSN (an important component of the extracellular actin-scavenging system) is responsible for the removal of actin that is released into the circulation during inflammation and from dying cells. We evaluated the levels of pGSN in plasma collected from groups of mice (5 mice in each) at 1 week (wk) and 1 month (1 mo) after exposure whole body to different doses of 28Si ions, i.e. 0, 0.1, 0.25, or 0.5 Gy (2 fractionated exposures, 15 days apart that totaled each selected dose). In the same mouse, the measurements of pGSN levels were coupled with the quantitation of injuries in the lung, determined by (a) the levels of cleaved poly (ADP-ribose) polymerase (cleaved-PARP), a marker of apoptotic cell-death, (b) the levels of activated nuclear factor-kappa B (NF-κB) and selected cytokines, i.e. tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), and IL-6, from tissue-lysates of the lung. Further, the ratio of neutrophils and lymphocytes (N/L) was determined in the same mouse. Our data indicated: (i) the magnitude of pGSN depletion was dependent to radiation dose at both harvest times, (ii) a persistent depletion of pGSN up to 1 mo post-exposure to 0.25 or 0.5 Gy of 28Si ions, (iii) an inverse-correlation between pGSN depletion and increased levels of cleaved-PARP, including activated NF-κB/pro-inflammatory cytokines in the lung, and (iv) at both harvest times, statistically significant increases in the N/L ratio in groups of mice exposed to 0.5 Gy only. Our findings suggested that depletion in pGSN levels reflects not only the responses to 28Si-ion exposure at both harvest times but also early and late-occurring damage.


Asunto(s)
Proteínas Sanguíneas/deficiencia , Gelsolina/deficiencia , Neumonía/sangre , Silicio/toxicidad , Oligoelementos/toxicidad , Animales , Proteínas Sanguíneas/efectos de la radiación , Muerte Celular , Gelsolina/sangre , Gelsolina/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos CBA , Neumonía/inducido químicamente , Neumonía/patología
12.
J Am Soc Nephrol ; 29(4): 1141-1153, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29335241

RESUMEN

The complement system is essential for host defense, but uncontrolled complement system activation leads to severe, mostly renal pathologies, such as atypical hemolytic uremic syndrome or C3 glomerulopathy. Here, we investigated a novel combinational approach to modulate complement activation by targeting C3 and the terminal pathway simultaneously. The synthetic fusion protein MFHR1 links the regulatory domains of complement factor H (FH) with the C5 convertase/C5b-9 inhibitory fragment of the FH-related protein 1. In vitro, MFHR1 showed cofactor and decay acceleration activity and inhibited C5 convertase activation and C5b-9 assembly, which prevented C3b deposition and reduced C3a/C5a and C5b-9 generation. Furthermore, this fusion protein showed the ability to escape deregulation by FH-related proteins and form multimeric complexes with increased inhibitory activity. In addition to substantially inhibiting alternative and classic pathway activation, MFHR1 blocked hemolysis mediated by serum from a patient with aHUS expressing truncated FH. In FH-/- mice, MFHR1 administration augmented serum C3 levels, reduced abnormal glomerular C3 deposition, and ameliorated C3 glomerulopathy. Taking the unique design of MFHR1 into account, we suggest that the combination of proximal and terminal cascade inhibition together with the ability to form multimeric complexes explain the strong inhibitory capacity of MFHR1, which offers a novel basis for complement therapeutics.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/sangre , Proteínas Sanguíneas/deficiencia , Proteínas Inactivadoras del Complemento C3b/genética , Inactivadores del Complemento/farmacología , Terapia Molecular Dirigida , Proteínas Recombinantes de Fusión/farmacología , Animales , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/inmunología , Complemento C3/metabolismo , Convertasas de Complemento C3-C5/antagonistas & inhibidores , Convertasas de Complemento C3-C5/metabolismo , Complemento C3b/antagonistas & inhibidores , Proteínas Inactivadoras del Complemento C3b/deficiencia , Complemento C5/metabolismo , Factor H de Complemento/genética , Inactivadores del Complemento/aislamiento & purificación , Inactivadores del Complemento/uso terapéutico , Complejo de Ataque a Membrana del Sistema Complemento/biosíntesis , Vía Alternativa del Complemento , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Glomérulos Renales/química , Glomérulos Renales/patología , Ratones , Ratones Noqueados , Dominios Proteicos , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/uso terapéutico
13.
Cell Death Dis ; 8(8): e3008, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28837146

RESUMEN

The limited efficacy of current treatment methods and increased type 2 diabetes mellitus (T2DM) incidence constitute an incentive for investigating how metabolic homeostasis is maintained, to improve treatment efficacy and identify novel treatment methods. We analyzed a three-generation family of Chinese origin with the common feature of T2DM attacks and fatty pancreas (FP), alongside 19 unrelated patients with FP and 58 cases with T2DM for genetic variations in Enho, serum adropin, and relative Treg amounts. Functional studies with adropin knockout (AdrKO) in C57BL/6J mice were also performed. It showed serum adropin levels were significantly lower in FP and T2DM patients than in healthy subjects; relative Treg amounts were also significantly decreased in FP and T2DM patients, and positively associated with adropin (r=0.7220, P=0.0001). Sequencing revealed that the patients shared a Cys56Trp mutation in Enho. In vivo, adropin-deficiency was associated with increased severity of glucose homeostasis impairment and fat metabolism disorder. AdrKO mice exhibited reduced endothelial nitric oxide synthase (eNOS) phosphorylation (Ser1177), impaired glycosphingolipid biosynthesis, adipocytes infiltrating, and loss of Treg, and developed FP and T2DM. Adropin-deficiency contributed to loss of Treg and the development of FP disease and T2DM.


Asunto(s)
Proteínas Sanguíneas/deficiencia , Diabetes Mellitus Tipo 2/terapia , Dieta Alta en Grasa/efectos adversos , Obesidad/complicaciones , Páncreas/patología , Péptidos/deficiencia , Animales , Diabetes Mellitus Tipo 2/patología , Femenino , Homeostasis , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Obesidad/patología
14.
Histochem Cell Biol ; 148(3): 219-227, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28421320

RESUMEN

Ubiquitin regulatory X domain-containing protein 8 (UBXD8) is engaged in the degradation of lipidated apolipoprotein B in hepatocytes. We previously showed that hepatocyte-specific UBXD8-deficient mice (U8-HKO) fed a moderately high-fat diet (31 kcal % fat) showed periportal macrovesicular steatosis along with a decrease in very low-density lipoprotein secretion, but did not develop fibrosis. In the present study, we examined whether U8-HKO mice show NASH-like phenotypes when fed a very high-fat diet (60 kcal % fat). U8-HKO mice and their age-matched littermates (control) were fed with two NASH model diets: choline-sufficient very high-fat diet and choline-deficient very high-fat diet. After being fed a very high-fat diet for 2 weeks, U8-HKO mice showed hepatic fibrosis in a significantly wider area than in the control. Fibrosis in U8-HKO mouse liver was further enhanced under a very high-fat diet depleted of choline (the liver surface was lumpy). Concomitant administration of an angiotensin 2 type 1 receptor blocker reduced the hepatic fibrosis caused by the very high-fat diet, suggesting the existence of inflammation. Carbon tetrachloride also induced hepatic fibrosis but the severity was comparable in the control and U8-HKO mice. In conjunction with our previous finding, the results indicate that although UBXD8 functionality can be largely compensated in the normal setting, it is crucial to sustain VLDL secretion when exposed to a dietary challenge of high fat. U8-HKO mice that develop fibrosis within 2 weeks of high-fat feeding can be used as a model to study NAFLD/NASH disease progression.


Asunto(s)
Proteínas Sanguíneas/deficiencia , Modelos Animales de Enfermedad , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Proteínas de la Membrana/deficiencia , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Proteínas Sanguíneas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados
15.
J Stroke Cerebrovasc Dis ; 25(9): e158-60, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27425767

RESUMEN

The authors describe herein the first adult case with moyamoya disease associated with protein Z deficiency. A 41-year-old woman was admitted to our hospital due to sudden onset of dysarthria and left extremity weakness 6 days after the delivery of her first baby. Previously, she repeated early fetal losses and was diagnosed with protein Z deficiency. Laboratory examination showed that the plasma concentration of protein Z was .73 µg/ml, being lower than the control. Radiological examination demonstrated typical findings of moyamoya disease with advanced stage on both sides. She successfully underwent surgical revascularization on both sides and was free from any cerebrovascular events during a follow-up period of 2 years. In addition to hemodynamic compromise, protein Z deficiency and hypercoagulation state after delivery might cause ischemic stroke in this case.


Asunto(s)
Proteínas Sanguíneas/deficiencia , Enfermedad de Moyamoya/complicaciones , Periodo Posparto/fisiología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/metabolismo , Adulto , Encéfalo/diagnóstico por imagen , Femenino , Humanos , Angiografía por Resonancia Magnética , Enfermedad de Moyamoya/diagnóstico por imagen , Accidente Cerebrovascular/diagnóstico por imagen
16.
Yakugaku Zasshi ; 136(5): 705-9, 2016.
Artículo en Japonés | MEDLINE | ID: mdl-27150923

RESUMEN

Although protein replacement is an effective treatment for serum protein deficiencies such as diabetes and hemophilia, recombinant protein products are not available for all rare inherited diseases due to the instability of the recombinant proteins and/or to cost. Gene therapy is the most attractive option for treating patients with such rare diseases. To develop an effective ex vivo gene therapy-based protein replacement treatment requires recipient cells that differ from those used in standard gene therapy, which is performed to correct the function of the recipient cells. Adipose tissue is an expected source of proliferative cells for cell-based therapies, including regenerative medicine and gene transfer applications. Based on recent advances in cell biology and extensive clinical experience in transplantation therapy for adipose tissue, we focused on the mature adipocyte fraction, which is the floating fraction after collagenase digestion and centrifugation of adipose tissue. Proliferative adipocytes were propagated from the floating fraction by the ceiling culture technique. These cells are designated as ceiling culture-derived proliferative adipocytes (ccdPAs). We first focused on lecithin:cholesterol acyltransferase (LCAT) deficiency, an inherited metabolic disorder caused by lcat gene mutation, and ccdPAs as a therapeutic gene vehicle for LCAT replacement therapy. In our recent in vitro and animal model studies, we developed an adipose cell manipulation procedure using advanced gene transduction methods and transplantation scaffolds. We herein introduce the progress made in novel adipose tissue-based therapeutic strategies for the treatment of protein deficiencies and describe their future applications for other intractable diseases.


Asunto(s)
Adipocitos , Terapia Genética/métodos , Terapia Genética/tendencias , Deficiencia de la Lecitina Colesterol Aciltransferasa/terapia , Transducción Genética/métodos , Adipocitos/trasplante , Animales , Proteínas Sanguíneas/deficiencia , Técnicas de Cultivo de Célula , Diabetes Mellitus/terapia , Modelos Animales de Enfermedad , Hemofilia A/terapia , Humanos , Ratones , Proteínas Recombinantes
17.
Arterioscler Thromb Vasc Biol ; 36(6): 1164-73, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27079884

RESUMEN

OBJECTIVE: Thiol isomerases facilitate protein folding in the endoplasmic reticulum, and several of these enzymes, including protein disulfide isomerase and ERp57, are mobilized to the surface of activated platelets, where they influence platelet aggregation, blood coagulation, and thrombus formation. In this study, we examined the synthesis and trafficking of thiol isomerases in megakaryocytes, determined their subcellular localization in platelets, and identified the cellular events responsible for their movement to the platelet surface on activation. APPROACH AND RESULTS: Immunofluorescence microscopy imaging was used to localize protein disulfide isomerase and ERp57 in murine and human megakaryocytes at various developmental stages. Immunofluorescence microscopy and subcellular fractionation analysis were used to localize these proteins in platelets to a compartment distinct from known secretory vesicles that overlaps with an inner cell-surface membrane region defined by the endoplasmic/sarcoplasmic reticulum proteins calnexin and sarco/endoplasmic reticulum calcium ATPase 3. Immunofluorescence microscopy and flow cytometry were used to monitor thiol isomerase mobilization in activated platelets in the presence and absence of actin polymerization (inhibited by latrunculin) and in the presence or absence of membrane fusion mediated by Munc13-4 (absent in platelets from Unc13d(Jinx) mice). CONCLUSIONS: Platelet-borne thiol isomerases are trafficked independently of secretory granule contents in megakaryocytes and become concentrated in a subcellular compartment near the inner surface of the platelet outer membrane corresponding to the sarco/endoplasmic reticulum of these cells. Thiol isomerases are mobilized to the surface of activated platelets via a process that requires actin polymerization but not soluble N-ethylmaleimide-sensitive fusion protein attachment receptor/Munc13-4-dependent vesicular-plasma membrane fusion.


Asunto(s)
Plaquetas/enzimología , Membrana Celular/enzimología , Megacariocitos/enzimología , Activación Plaquetaria , Proteína Disulfuro Isomerasas/sangre , Actinas/sangre , Animales , Plaquetas/efectos de los fármacos , Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/genética , Calnexina/sangre , Membrana Celular/efectos de los fármacos , Genotipo , Humanos , Megacariocitos/efectos de los fármacos , Fusión de Membrana , Proteínas de la Membrana/sangre , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Activación Plaquetaria/efectos de los fármacos , Proteína Disulfuro Isomerasas/biosíntesis , Transporte de Proteínas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/sangre
18.
Surg Obes Relat Dis ; 12(3): 629-634, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27012874

RESUMEN

BACKGROUND: Electrolyte and nutritional deficiencies have been reported in Western populations seeking bariatric surgery. However, data are scarce for Chinese patients. OBJECTIVES: To investigate the prevalence of electrolyte and nutritional deficiencies in Chinese bariatric surgery candidates and to explore their associations with patients' demographic data. SETTING: University hospital, China. METHODS: Demographical data of 211 patients presenting for bariatric surgery were collected on gender, age, body mass index (BMI) and waist circumference (WC). Blood biochemical data were collected on some nutrients (hemoglobin, albumin, globulin, folate, vitamin B12, calcium, phosphorus, iron, ferritin, magnesium, parathyroid hormone [PTH], and vitamin D) and some electrolytes (potassium, sodium, and chloride). RESULTS: Deficiencies were found for hemoglobin (2.8%), albumin (11.8%), globulin (1.4%), folate (32.2%), vitamin B12 (4.7%), corrected calcium (13.7%), phosphorus (10.4%), iron (9.0%), ferritin (1.9%), vitamin D (80.0%), potassium (5.7%), sodium (7.6%), and chloride (15.6%). Secondary hyperparathyroidism was found in 17.3%; no hypomagnesemia was encountered. A significant correlation was observed between age and folate, corrected calcium and PTH levels (r = .257, -.206, and .273, respectively; P<.05). Greater BMI was associated with lower albumin and folate (r = -.338 and -.370, respectively) and with higher globulin and phosphorus levels (r = .267 and .138, respectively). Folate deficiency was more common in the 18- to 30-year-old age group (P = .042) and the patients with BMI>45 kg/m(2) (P = .001). WC had an association with rates of albumin, folate, and corrected calcium deficiencies, as well as hemoglobin, albumin, and globulin, folate, phosphorus, and ferritin levels. CONCLUSION: Electrolyte and nutritional deficiencies are common in Chinese bariatric surgery candidates. Routine evaluation of electrolyte and nutritional levels should be carried out in this population.


Asunto(s)
Cirugía Bariátrica , Enfermedades Carenciales/etiología , Electrólitos/metabolismo , Adolescente , Adulto , Anciano , Avitaminosis/etnología , Avitaminosis/etiología , Proteínas Sanguíneas/deficiencia , Índice de Masa Corporal , China/etnología , Enfermedades Carenciales/etnología , Femenino , Deficiencia de Ácido Fólico/etnología , Deficiencia de Ácido Fólico/etiología , Humanos , Masculino , Micronutrientes/deficiencia , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/etnología , Obesidad/cirugía , Estudios Retrospectivos , Circunferencia de la Cintura/etnología , Circunferencia de la Cintura/fisiología , Adulto Joven
20.
Int J Clin Exp Pathol ; 7(9): 6064-71, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25337252

RESUMEN

BACKGROUND: Protein Z (PZ) is a vitamin K-dependent coagulation factor without catalytic activity. Evidence points towards PZ as an independent risk factor for the occurrence of human atherosclerotic vascular diseases. The aim of this study was to investigate the role of PZ in vascular arterial disease. MATERIAL AND METHODS: PZ-deficient (PZ(-/-)) mice and their wild-type littermates (PZ(+/+)) were subjected to unilateral carotid artery injury by using ferric chloride and dissected 21 days thereafter for histological analysis. Human aortic smooth muscle cells (SMC) were used for in vitro wound healing assay to assess the influence of PZ on SMC migration and for cell proliferation studies. RESULTS: Morphometric analysis of neointima formation revealed a significantly increased area and thickness of the neointima and subsequently increased luminal stenosis in carotid arteries of PZ(-/-) mice compared to PZ(+/+) mice (p < 0.05, n = 9). Immunohistochemical analysis of neointima lesion composition revealed significantly higher numbers of PCNA-positive and α-SMA-positive cells in the neointima of PZ(-/-) mice. Furthermore, PZ showed an anti-migratory potency in in vitro wound healing assay with SMCs, while no effect of PZ on SMC proliferation was detectable. Conclusion: PZ contributes to a reduced neointima formation after vascular injury, underlining the modulatory role of the coagulation cascade in vascular homeostasis.


Asunto(s)
Proteínas Sanguíneas/deficiencia , Traumatismos de las Arterias Carótidas/metabolismo , Inflamación/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima , Lesiones del Sistema Vascular/metabolismo , Actinas/metabolismo , Animales , Proteínas Sanguíneas/genética , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/inducido químicamente , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Cloruros , Modelos Animales de Enfermedad , Compuestos Férricos , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factores de Tiempo , Lesiones del Sistema Vascular/inducido químicamente , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...