Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 611
Filtrar
1.
Bioconjug Chem ; 35(5): 653-664, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38593046

RESUMEN

Disorder of complement response is a significant pathogenic factor causing some autoimmune and inflammation diseases. The Ornithodoros moubata Complement Inhibitor (OmCI), a small 17 kDa natural protein, was initially extracted from soft tick salivary glands. The protein was found binding to complement C5 specifically, inhibiting the activation of the complement pathway, which is a successful therapeutic basis of complement-mediated diseases. However, a short half-life due to rapid renal clearance is a common limitation of small proteins for clinical application. In this study, we extended the half-life of OmCI by modifying it with fatty acid, which was a method used to improve the pharmacokinetics of native peptides and proteins. Five OmCI mutants were initially designed, and single-site cysteine mutation was introduced to each of them. After purification, four OmCI mutants were obtained that showed similar in vitro biological activities. Three mutants of them were subsequently coupled with different fatty acids by nucleophilic substitution. In total, 15 modified derivatives were screened and tested for anticomplement activity in vitro. The results showed that coupling with fatty acid would not significantly affect their complement-inhibitory activity (CH50 and AH50). OmCIT90C-CM02 and OmCIT90C-CM05 were validated as the applicable OmCI bioconjugates for further pharmacokinetic assessments, and both showed improved plasma half-life in mice compared with unmodified OmCI (15.86, 17.96 vs 2.57 h). In summary, our data demonstrated that OmCI conjugated with fatty acid could be developed as the potential long-acting C5 complement inhibitor in the clinic.


Asunto(s)
Complemento C5 , Ácidos Grasos , Ornithodoros , Animales , Ácidos Grasos/química , Ratones , Complemento C5/antagonistas & inhibidores , Diseño de Fármacos , Semivida , Proteínas Inactivadoras de Complemento/farmacología , Proteínas Inactivadoras de Complemento/química , Inactivadores del Complemento/farmacología , Inactivadores del Complemento/farmacocinética , Inactivadores del Complemento/química , Humanos
2.
Phytomedicine ; 107: 154453, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36116199

RESUMEN

BACKGROUND: Owing to the involvement of the overactivated complement system in acute lung injury (ALI) development, anticomplement components may attenuate ALI. Hedyotis diffusa is a traditional Chinese medicine for treating lung heat and its crude polysaccharides (HDP) exhibit significant anticomplement activity in vitro. PURPOSE: To obtain an anticomplement homogeneous polysaccharide from HDP and verify its therapeutic effect and mechanism on ALI. METHODS: Diethylaminoethyl-52 (DEAE-52) cellulose and gel permeation columns were used to isolate a homogeneous polysaccharide HD-PS-3, which was then characterized using nuclear magnetic resonance (NMR) and methylation analysis. In vitro, the anticomplement activities of HD-PS-3 through classical and alternative pathways were determined using a hemolytic test. The therapeutic effects of HDP and HD-PS-3 on ALI were evaluated in lipopolysaccharide (LPS) intratracheal instilled mice. Hematoxylin and eosin (H&E) staining, enzyme-linked immunosorbent assay (ELISA), and immunohistochemical staining were used to assess histological changes, measure cytokine levels, and evaluate the degree of complement component 3c (C3c) deposition and neutrophil infiltration, respectively. ELISA, western blotting, and immunofluorescence were used to analyze neutrophil extracellular trap (NET) formation. RESULTS: From HDP, 1.5 g of the homogeneous polysaccharide HD-PS-3 was obtained. HD-PS-3 was an acidic heteropolysaccharide with an acetyl group, which was composed of →4,6)-α-Glcp-(1→, →3,4)-α-Glcp-(1→, →4)-α-Glcp-(1→, →4,6)-α-Galp-(1→, →5)-α-Araf-(1→, α-Rhap-(1→, α-Araf-(1→, α-GlcpA-(1→, →4)-ß-Manp-(1→, ß-Manp-(1→ and →3)-ß-Manp-(1→. The in vitro results suggest that HD-PS-3 exhibited anticomplement activity with CH50 and AP50 values of 115 ± 12 µg/ml and 307 ± 11 µg/ml, respectively. After confirming the efficacy of HDP (200 mg/kg) in attenuating lung injury, the effect of HD-PS-3 on ALI was also investigated. HD-PS-3 (75 and 150 mg/kg) attenuated LPS-induced ALI as well, evidenced by lung pathology, lung injury scores, protein concentration, leukocyte counts, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) contents in bronchoalveolar lavage fluid (BALF). Mechanistically, HD-PS-3 inhibited complement activation, manifested in reduced pulmonary C3c deposition in lung tissue and complement component 3a (C3a) content in BALF. Neutrophil recruitment was also reduced by HD-PS-3, with significantly reduced pulmonary neutrophil infiltration and lower levels of C-X-C motif chemokine ligand 1 (CXCL1) and myeloperoxidase (MPO) in BALF. In addition, HD-PS-3 reduced the levels of MPO-DNA complex in BALF, decreased citrullinated histone H3 (Cit H3) expression and NET formation (colocalization of MPO, Cit H3, and DNA) in lung tissue. CONCLUSION: An anticomplement homogeneous polysaccharide HD-PS-3 was isolated from H. diffusa. HD-PS-3 exhibited a therapeutic effect against ALI, and the mechanism might be related to its inhibitory effects on complement activation, neutrophil recruitment, and NET formation.


Asunto(s)
Lesión Pulmonar Aguda , Trampas Extracelulares , Hedyotis , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Animales , Quimiocinas/metabolismo , Complemento C3a/metabolismo , Complemento C3c/metabolismo , Proteínas Inactivadoras de Complemento , Citocinas/metabolismo , Trampas Extracelulares/metabolismo , Histonas , Interleucina-6/metabolismo , Ligandos , Lipopolisacáridos , Ratones , Peroxidasa/metabolismo , Polisacáridos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
3.
J Clin Immunol ; 40(5): 699-707, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32447592

RESUMEN

Hemophagocytic lymphohistiocytosis (HLH) is a syndrome of excessive immune system activation driven mainly by high levels of interferon gamma. The clinical presentation of HLH can have considerable overlap with other inflammatory conditions. We present a cohort of patients with therapy refractory HLH referred to our center who were found to have a simultaneous presentation of complement-mediated thrombotic microangiopathy (TMA). Twenty-three patients had therapy refractory HLH (13 primary, 4 EVB-HLH, 6 HLH without known trigger). Sixteen (69.6%) met high-risk TMA criteria. Renal failure requiring renal replacement therapy, severe hypertension, serositis, and gastrointestinal bleeding were documented only in patients with HLH who had concomitant complement-mediated TMA. Patients with HLH and without TMA required ventilator support mainly due to CNS symptoms, while those with HLH and TMA had respiratory failure predominantly associated with pulmonary hypertension, a known presentation of pulmonary TMA. Ten patients received eculizumab for complement-mediated TMA management while being treated for HLH. All patients who received the complement blocker eculizumab in addition to the interferon gamma blocker emapalumab had complete resolution of their TMA and survived. Our observations suggest co-activation of both interferon and complement pathways as a potential culprit in the evolution of thrombotic microangiopathy in patients with inflammatory disorders like refractory HLH and may offer novel therapeutic approaches for these critically ill patients. TMA should be considered in children with HLH and multi-organ failure, as an early institution of a brief course of complement blocking therapy in addition to HLH-targeted therapy may improve clinical outcomes in these patients.


Asunto(s)
Interferón gamma/metabolismo , Linfohistiocitosis Hemofagocítica/inmunología , Microangiopatías Trombóticas/inmunología , Adolescente , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Niño , Preescolar , Estudios de Cohortes , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento/metabolismo , Femenino , Humanos , Hipertensión Pulmonar , Lactante , Interferón gamma/antagonistas & inhibidores , Linfohistiocitosis Hemofagocítica/mortalidad , Masculino , Insuficiencia Respiratoria , Microangiopatías Trombóticas/mortalidad , Adulto Joven
4.
Pediatr Nephrol ; 35(1): 153-162, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31667615

RESUMEN

BACKGROUND: C3 glomerulopathy (C3G) is defined by dominant glomerular deposition of C3 and minimal or no immunoglobulin, with two subtypes-dense deposit disease (DDD) and C3 glomerulonephritis (C3GN)-distinguished by features on electron microscopy (EM). Given that this rare disease has generally unfavorable yet highly variable outcomes, we sought out to review the histopathology, complement/genetic studies, and renal outcomes of pediatric patients with C3G at our institution. METHODS: All native kidney biopsies performed in a single pediatric hospital over a 10-year period were reviewed for features of C3G. Of 589 biopsy reports, we identified 9 patients fulfilling the diagnostic criteria for C3G and retrospectively reviewed their clinical chart and renal biopsy findings. RESULTS: We identified 4 patients with DDD, 4 with C3GN, and 1 indeterminate case, with features of both C3GN and DDD. Five patients were positive for one or more nephritic factors (C3NeF, C4NeF, C5NeF) with 1 patient additionally positive for complement factor H (CFH) autoantibody. Genetic testing done in 5 of the 9 patients failed to identify any causative mutations. Three patients showed progressive renal dysfunction over a mean follow-up period of 33 months. CONCLUSIONS: Complement and genetic studies are now routinely recommended for patients with a histopathological diagnosis of C3G. Careful interpretation of these studies and their prognostic and therapeutic implications in conjunction with biopsy findings is needed to further understand the pathophysiology of this rare disease in children.


Asunto(s)
Complemento C3/inmunología , Glomerulonefritis Membranoproliferativa/inmunología , Fallo Renal Crónico/epidemiología , Glomérulos Renales/patología , Adolescente , Biopsia , Niño , Complemento C3/genética , Proteínas Inactivadoras de Complemento/análisis , Proteínas Inactivadoras de Complemento/inmunología , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Pruebas Genéticas , Glomerulonefritis Membranoproliferativa/sangre , Glomerulonefritis Membranoproliferativa/complicaciones , Glomerulonefritis Membranoproliferativa/genética , Humanos , Fallo Renal Crónico/inmunología , Glomérulos Renales/inmunología , Glomérulos Renales/ultraestructura , Masculino , Microscopía Electrónica , Mutación , Estudios Retrospectivos
5.
Am J Chin Med ; 47(4): 823-839, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31091972

RESUMEN

Ginseng root has been used in traditional oriental medicine for the enhancement of immune system function. The immunostimulatory effects of ginseng berry polysaccharides, however, remain unclear. Effects of polysaccharides from ginseng berry on the activation of natural killer (NK) cells and inhibition of tumors are reported. A crude polysaccharide was isolated from ginseng berry as a ginseng berry polysaccharide portion (GBPP) and was further fractionated using gel filtration chromatography to obtain the three polysaccharide fractions GBPP-I, -II and -III. GBPP-I consisted of mainly galactose (46.9%) and arabinose (27.5%). GBPP-I showed a high dose-dependent anticomplementary activity. Stimulation of murine peritoneal macrophages by GBPP-I showed the greatest enhancement of interleukin (IL)-6 and IL-12 and tumor necrosis factor (TNF)- α production. In addition, an ex vivo assay of natural killer (NK) cell activity showed that oral ( p.o.) administration of GBPP-I significantly increased NK cell cytotoxicity in YAC-1 tumor cells and production of granzyme B. Prophylactic intravenous ( i.v.) and p.o. administration of GBPP-I significantly and dose-dependently inhibited lung metastatic activity in B16BL6 melanoma cells. Depletion of NK cells after injection of rabbit anti-asialo GM1 partially abolished the inhibitory effect of GBPP-I on lung metastasis, indicating that NK cells play an important role in anticancer effects. GBPP-I exerts a strong immune-enhancing activity and can prevent cancer metastasis through activation of NK cells and other immune-related cells.


Asunto(s)
Adyuvantes Inmunológicos , Proteínas Inactivadoras de Complemento , Frutas/química , Macrófagos Peritoneales/inmunología , Panax/química , Polisacáridos/aislamiento & purificación , Polisacáridos/farmacología , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Células Asesinas Naturales/inmunología , Macrófagos Peritoneales/metabolismo , Melanoma Experimental/patología , Ratones Endogámicos BALB C , Polisacáridos/administración & dosificación , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
6.
Front Immunol ; 10: 752, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024572

RESUMEN

The interactions of cancer cells with components of the complement system are highly complex, leading to an outcome that is either favorable or detrimental to cancer cells. Currently, we perceive only the "tip of the iceberg" of these interactions. In this review, we focus on the complement terminal C5b-9 complex, known also as the complement membrane attack complex (MAC) and discuss the complexity of its interaction with cancer cells, starting with a discussion of its proposed mode of action in mediating cell death, and continuing with a portrayal of the strategies of evasion exhibited by cancer cells, and closing with a proposal of treatment approaches targeted at evasion strategies. Upon intense complement activation and membrane insertion of sufficient C5b-9 complexes, the afflicted cells undergo regulated necrotic cell death with characteristic damage to intracellular organelles, including mitochondria, and perforation of the plasma membrane. Several pro-lytic factors have been proposed, including elevated intracellular calcium ion concentrations and activated JNK, Bid, RIPK1, RIPK3, and MLKL; however, further research is required to fully characterize the effective cell death signals activated by the C5b-9 complexes. Cancer cells over-express a multitude of protective measures which either block complement activation, thus reducing the number of membrane-inserted C5b-9 complexes, or facilitate the elimination of C5b-9 from the cell surface. Concomitantly, cancer cells activate several protective pathways that counteract the death signals. Blockage of complement activation is mediated by the complement membrane regulatory proteins CD46, CD55, and CD59 and by soluble complement regulators, by proteases that cleave complement proteins and by protein kinases, like CK2, which phosphorylate complement proteins. C5b-9 elimination and inhibition of cell death signals are mediated by caveolin and dynamin, by Hsp70 and Hsp90, by the mitochondrial stress protein mortalin, and by the protein kinases PKC and ERK. It is conceivable that various cancers and cancers at different stages of development will utilize distinct patterns of these and other MAC resistance strategies. In order to enhance the impact of antibody-based therapy on cancer, novel precise reagents that block the most effective protective strategies will have to be designed and applied as adjuvants to the therapeutic antibodies.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Neoplasias/inmunología , Animales , Señalización del Calcio , Muerte Celular/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Activación de Complemento , Proteínas Inactivadoras de Complemento/inmunología , Proteínas Inactivadoras de Complemento/metabolismo , Citotoxicidad Inmunológica , Humanos , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Modelos Inmunológicos , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral/inmunología
7.
J Am Soc Nephrol ; 29(2): 620-635, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29042454

RESUMEN

Complement-activating anti-HLA donor-specific antibodies (DSAs) are associated with impaired kidney transplant outcome; however, whether these antibodies induce a specific rejection phenotype and influence response to therapy remains undetermined. We prospectively screened 931 kidney recipients for complement-activating DSAs and used histopathology, immunostaining, and allograft gene expression to assess rejection phenotypes. Effector cells were evaluated using in vitro human cell cultures. Additionally, we assessed the effect of complement inhibition on kidney allograft rejection phenotype and the clinical response to complement inhibition in 116 independent kidney recipients with DSAs at transplant receiving rejection prophylaxis with eculizumab or standard of care (plasma exchange and intravenous Ig) at ten international centers. The histomolecular rejection phenotype associated with complement-activating DSA was characterized by complement deposition and accumulation of natural killer cells and monocytes/macrophages in capillaries and increased expression of five biologically relevant genes (CXCL11, CCL4, MS4A7, MS4A6A, and FCGR3A) indicative of endothelial activation, IFNγ response, CD16-mediated natural killer cell activation, and monocyte/macrophage activation. Compared with standard of care, eculizumab specifically abrogated this histomolecular rejection phenotype and associated with a decreased 3-month rejection incidence rate in patients with complement-activating DSAs (56%; 95% confidence interval [95% CI], 38% to 74% versus 19%; 95% CI, 8% to 35%; P=0.001) but not in those with noncomplement-activating DSAs (9%; 95% CI, 2% to 25% versus 13%; 95% CI, 2% to 40%; P=0.65). In conclusion, circulating complement-activating anti-HLA DSAs are associated with a specific histomolecular kidney allograft rejection phenotype that can be abrogated by complement inhibition.


Asunto(s)
Aloinjertos/inmunología , Anticuerpos/sangre , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Antígenos HLA/inmunología , Transcriptoma , Adulto , Anciano , Aloinjertos/metabolismo , Aloinjertos/patología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Células Cultivadas , Quimiocina CCL4/genética , Quimiocina CXCL11/genética , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento/metabolismo , Femenino , Rechazo de Injerto/terapia , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Factores Inmunológicos/uso terapéutico , Riñón/patología , Trasplante de Riñón , Células Asesinas Naturales , Macrófagos , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Monocitos , Fenotipo , Intercambio Plasmático , Receptores de IgG/genética
8.
Nat Rev Dis Primers ; 3: 17028, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28516949

RESUMEN

Paroxysmal nocturnal haemoglobinuria (PNH) is a clonal haematopoietic stem cell (HSC) disease that presents with haemolytic anaemia, thrombosis and smooth muscle dystonias, as well as bone marrow failure in some cases. PNH is caused by somatic mutations in PIGA (which encodes phosphatidylinositol N-acetylglucosaminyltransferase subunit A) in one or more HSC clones. The gene product of PIGA is required for the biosynthesis of glycosylphosphatidylinositol (GPI) anchors; thus, PIGA mutations lead to a deficiency of GPI-anchored proteins, such as complement decay-accelerating factor (also known as CD55) and CD59 glycoprotein (CD59), which are both complement inhibitors. Clinical manifestations of PNH occur when a HSC clone carrying somatic PIGA mutations acquires a growth advantage and differentiates, generating mature blood cells that are deficient of GPI-anchored proteins. The loss of CD55 and CD59 renders PNH erythrocytes susceptible to intravascular haemolysis, which can lead to thrombosis and to much of the morbidity and mortality of PNH. The accumulation of anaphylatoxins (such as C5a) from complement activation might also have a role. The natural history of PNH is highly variable, ranging from quiescent to life-threatening. Therapeutic strategies include terminal complement blockade and bone marrow transplantation. Eculizumab, a monoclonal antibody complement inhibitor, is highly effective and the only licensed therapy for PNH.


Asunto(s)
Hemoglobinuria Paroxística/patología , Hemoglobinuria Paroxística/terapia , Proteínas de la Membrana/genética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Trasplante de Médula Ósea , Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Proteínas Inactivadoras de Complemento/metabolismo , Hemoglobinuria Paroxística/genética , Hemoglobinuria Paroxística/metabolismo , Humanos , Mutación , Resultado del Tratamiento
9.
J Intern Med ; 282(2): 116-128, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28345259

RESUMEN

The complement system is an arm of innate immunity that aids in the removal of pathogens and dying cells. Due to its harmful, pro-inflammatory potential, complement is controlled by several soluble and membrane-bound inhibitors. This family of complement regulators has been recently extended by the discovery of several new members, and it is becoming apparent that these proteins harbour additional functions. In this review, the current state of knowledge of the physiological functions of four complement regulators will be described: cartilage oligomeric matrix protein (COMP), CUB and sushi multiple domains 1 (CSMD1), sushi domain-containing protein 4 (SUSD4) and CD59. Complement activation is involved in both the development of and defence against cancer. COMP expression is pro-oncogenic, whereas CSMD1 and SUSD4 act as tumour suppressors. These effects may be related in part to the complex influence of complement on cancer but also depend on unrelated functions such as the protection of cells from endoplasmic reticulum stress conveyed by intracellular COMP. CD59 is the main inhibitor of the membrane attack complex, and its deficiency leads to complement attack on erythrocytes and severe haemolytic anaemia, which is now amenable to treatment with an inhibitor of C5 cleavage. Unexpectedly, the intracellular pool of CD59 is crucial for insulin secretion from pancreatic ß-cells. This finding is one of several relating to the intracellular functions of complement proteins, which until recently were only considered to be present in the extracellular space. Understanding the alternative functions of complement inhibitors may unravel unexpected links between complement and other physiological systems, but is also important for better design of therapeutic complement inhibition.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Animales , Antígenos CD59/fisiología , Proteína de la Matriz Oligomérica del Cartílago/fisiología , Activación de Complemento/fisiología , Proteínas Inactivadoras de Complemento/fisiología , Humanos , Infecciones/fisiopatología , Inflamación/fisiopatología , Artropatías/fisiopatología , Proteínas de la Membrana/fisiología , Neoplasias/fisiopatología , Proteínas Supresoras de Tumor
10.
J Gen Virol ; 97(8): 1765-1770, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27153814

RESUMEN

Newcastle disease virus (NDV) is being developed as an oncolytic virus for virotherapy. In this study we analysed the regulation of complement-mediated inactivation of a recombinant NDV in different host cells. NDV grown in human cells was less sensitive to complement-mediated virus inactivation than NDV grown in embryonated chicken eggs. Additionally, NDV produced from HeLa-S3 cells is more resistant to complement than NDV from 293F cells, which correlated with higher expression and incorporation of complement regulatory proteins (CD46, CD55 and CD59) into virions from HeLa-S3 cells. Further analysis of the recombinant NDVs individually expressing the three CD molecules showed that CD55 is the most potent in counteracting complement-mediated virus inactivation. The results provide important information on selecting NDV manufacture substrate to mitigate complement-mediated virus inactivation.


Asunto(s)
Antígenos CD55/metabolismo , Proteínas Inactivadoras de Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Interacciones Huésped-Patógeno , Factores Inmunológicos/metabolismo , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/fisiología , Animales , Antígenos CD59/metabolismo , Línea Celular , Pollos , Humanos , Proteína Cofactora de Membrana/metabolismo
12.
BMC Cancer ; 15: 737, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26480818

RESUMEN

BACKGROUND: The human Sushi Domain-Containing Protein 4 (SUSD4) was recently shown to function as a novel inhibitor of the complement system, but its role in tumor progression is unknown. METHODS: Using immunohistochemistry and quantitative PCR, we investigated SUSD4 expression in breast cancer tissue samples from two cohorts. The effect of SUSD4 expression on cell migration and invasion was studied in vitro using two human breast cancer cell lines overexpressing SUSD4. RESULTS: Tissue stainings revealed that both tumor cells and tumor-infiltrating cells expressed SUSD4. The highest SUSD4 expression was detected in differentiated tumors with decreased rate of metastasis, and SUSD4 expression was associated with improved survival of the patients. Moreover, forced SUSD4 expression in human breast cancer cells attenuated their migratory and invasive traits in culture. SUSD4 expression also inhibited colony formation of human breast cancer cells cultured on carcinoma-associated fibroblasts. Furthermore, large numbers of SUSD4-expressing T cells in the tumor stroma associated with better overall survival of the breast cancer patients. CONCLUSION: Our findings indicate that SUSD4 expression in both breast cancer cells and T cells infiltrating the tumor-associated stroma is useful to predict better prognosis of breast cancer patients.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Inactivadoras de Complemento/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/genética , ARN Neoplásico/genética , Linfocitos T/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proteínas Inactivadoras de Complemento/biosíntesis , Femenino , Humanos , Inmunohistoquímica , Proteínas de la Membrana/biosíntesis , Microscopía Fluorescente , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Pronóstico , Linfocitos T/patología
13.
Biochem Soc Trans ; 43(5): 812-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26517887

RESUMEN

Complement control protein modules (CCPs) occur in numerous functionally diverse extracellular proteins. Also known as short consensus repeats (SCRs) or sushi domains each CCP contains approximately 60 amino acid residues, including four consensus cysteines participating in two disulfide bonds. Varying in length and sequence, CCPs adopt a ß-sandwich type fold and have an overall prolate spheroidal shape with N- and C-termini lying close to opposite poles of the long axis. CCP-containing proteins are important as cytokine receptors and in neurotransmission, cell adhesion, blood clotting, extracellular matrix formation, haemoglobin metabolism and development, but CCPs are particularly well represented in the vertebrate complement system. For example, factor H (FH), a key soluble regulator of the alternative pathway of complement activation, is made up entirely from a chain of 20 CCPs joined by short linkers. Collectively, therefore, the 20 CCPs of FH must mediate all its functional capabilities. This is achieved via collaboration and division of labour among these modules. Structural studies have illuminated the dynamic architectures that allow FH and other CCP-rich proteins to perform their biological functions. These are largely the products of a highly varied set of intramolecular interactions between CCPs. The CCP can act as building block, spacer, highly versatile recognition site or dimerization mediator. Tandem CCPs may form composite binding sites or contribute to flexible, rigid or conformationally 'switchable' segments of the parent proteins.


Asunto(s)
Enzimas Activadoras de Complemento/química , Activación de Complemento , Proteínas Inactivadoras de Complemento/química , Diseño de Fármacos , Modelos Moleculares , Ingeniería de Proteínas , Animales , Sitios de Unión , Enzimas Activadoras de Complemento/genética , Enzimas Activadoras de Complemento/metabolismo , Factor H de Complemento/química , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Inactivadores del Complemento/química , Inactivadores del Complemento/metabolismo , Inactivadores del Complemento/farmacología , Proteínas Inactivadoras de Complemento/genética , Proteínas Inactivadoras de Complemento/metabolismo , Humanos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
14.
J Immunol ; 195(3): 1129-38, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26071558

RESUMEN

Defective placentation and subsequent placental insufficiency lead to maternal and fetal adverse pregnancy outcome, but their pathologic mechanisms are unclear, and treatment remains elusive. The mildly hypertensive BPH/5 mouse recapitulates many features of human adverse pregnancy outcome, with pregnancies characterized by fetal loss, growth restriction, abnormal placental development, and defects in maternal decidual arteries. Using this model, we show that recruitment of neutrophils triggered by complement activation at the maternal/fetal interface leads to elevation in local TNF-α levels, reduction of the essential angiogenic factor vascular endothelial growth factor, and, ultimately, abnormal placentation and fetal death. Blockade of complement with inhibitors specifically targeted to sites of complement activation, depletion of neutrophils, or blockade of TNF-α improves spiral artery remodeling and rescues pregnancies. These data underscore the importance of innate immune system activation in the pathogenesis of placental insufficiency and identify novel methods for treatment of pregnancy loss mediated by abnormal placentation.


Asunto(s)
Aborto Espontáneo/prevención & control , Activación de Complemento/inmunología , Inmunidad Innata , Neutrófilos/inmunología , Insuficiencia Placentaria/prevención & control , Placentación/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Aborto Espontáneo/inmunología , Animales , Línea Celular , Proteínas Inactivadoras de Complemento/farmacología , Proteínas del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Femenino , Muerte Fetal , Retardo del Crecimiento Fetal/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/inmunología , Placenta/citología , Insuficiencia Placentaria/inmunología , Embarazo , Trofoblastos/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Pediatr Blood Cancer ; 62(2): 224-228, 2015 02.
Artículo en Inglés | MEDLINE | ID: mdl-25382742

RESUMEN

BACKGROUND: Addition of anti-GD2 antibody ch14.18 to the treatment of neuroblastoma has improved outcomes. The most common side effect of ch14.18 is neuropathic pain, which may in part be complement-mediated. Hu14.18K322A is a humanized anti-GD2 antibody designed to diminish complement activation and induce less pain. We compare the pain outcomes in patients treated with ch14.18 and those treated with hu14.18K322A, and explore dose-dependent relationships between pain scores, opioid requirements, and complement levels in patients treated with hu14.18K322A. PROCEDURE: Opioid (morphine equivalent mg/kg) and anxiolytic requirements during course 1 (4 days) in patients treated with hu14.18K322A and ch14.18 were reviewed. Correlations between antibody dose and pain scores, opioid requirements, and complement levels were examined for patients receiving hu14.18K322A. RESULTS: Patients treated with hu14.18K322A (n = 19) had lower opioid requirements than those who received ch14.18 (n = 9). The differences in median opioid requirements (mg/kg) were statistically significant for the overall course (1.57 vs. 2.41, P = 0.019) as well as for Days 3 (0.34 vs. 0.65, P = 0.005), and 4 (0.32 vs. 0.64, P = 0.010). No difference in anxiolytic use was observed between the two groups. In the group treated with hu14.18K322A, we found a positive correlation between antibody dose administered and pain scores, but no correlation between antibody dose and opioid requirements or changes in complement levels. CONCLUSIONS: In this retrospective analysis, hu14.18K322A induced less pain than ch14.18 based on opioid requirements. Pediatr Blood Cancer 2015;62:224-228. © 2014 Wiley Periodicals, Inc.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Proteínas Inactivadoras de Complemento/uso terapéutico , Gangliósidos/antagonistas & inhibidores , Inmunoterapia/métodos , Neuralgia/tratamiento farmacológico , Neuroblastoma/terapia , Adolescente , Niño , Preescolar , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/efectos de los fármacos , Femenino , Gangliósidos/inmunología , Humanos , Lactante , Masculino , Estudios Retrospectivos , Resultado del Tratamiento
16.
Clin Exp Immunol ; 177(2): 500-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24730624

RESUMEN

While there is evidence of a pathogenic role for complement in inflammatory bowel disease, there is also evidence for a protective role that relates to host defence and protection from endotoxaemia. There is thus concern regarding the use of systemic complement inhibition as a therapeutic strategy. Local delivery of a complement inhibitor to the colon by oral administration would ameliorate such concerns, but while formulations exist for oral delivery of low molecular weight drugs to the colon, they have not been used successfully for oral delivery of proteins. We describe a novel pellet formulation consisting of cross-linked dextran coated with an acrylic co-polymer that protects the complement inhibitor CR2-Crry from destruction in the gastrointestinal tract. CR2-Crry containing pellets administered by gavage, were characterized using a therapeutic protocol in a mouse model of dextran sulphate sodium (DSS)-induced colitis. Oral treatment of established colitis over a 5-day period significantly reduced mucosal inflammation and injury, with similar therapeutic benefit whether or not the proton pump inhibitor, omeprazole, was co-administered. Reduction in injury was associated with the targeting of CR2-Crry to the mucosal surface and reduced local complement activation. Treatment had no effect on systemic complement activity. This novel method for oral delivery of a targeted protein complement inhibitor will reduce systemic effects, thereby decreasing the risk of opportunistic infection, as well as lowering the required dose and treatment cost and improving patient compliance. Furthermore, the novel delivery system described here may provide similar benefits for administration of other protein-based drugs, such as anti-tumour necrosis factor-α antibodies.


Asunto(s)
Colitis/inmunología , Colon/efectos de los fármacos , Colon/inmunología , Proteínas Inactivadoras de Complemento/administración & dosificación , Proteínas del Sistema Complemento/inmunología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Administración Oral , Animales , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colon/patología , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/patología , Ratones , Proteínas Recombinantes de Fusión/administración & dosificación
17.
Gene Ther ; 21(5): 507-13, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24670995

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness among the elderly. Approximately 50% of AMD patients have a polymorphism in the negative regulator of complement known as Factor H. Individuals homozygous for a Y402H polymorphism in Factor H have elevated levels of membrane attack complex (MAC) in their choroid and retinal pigment epithelium relative to individuals homozygous for the wild-type allele. An inability to form MAC due to a polymorphism in C9 is protective against the formation of choroidal neovascularization (CNV) in AMD patients. Hence, blocking MAC in AMD patients may be protective against CNV. Here we investigate the potential of human proline/arginine-rich end leucine-rich repeat protein (PRELP) as an inhibitor of complement-mediated damage when delivered via the subretinal route using an AAV2/8 vector. In a fluorescence-activated cell sorting (FACS) lysis assay, PRELP inhibited normal human serum-mediated lysis of Hepa-1c1c7 cells by 18.7%. Unexpectedly, PRELP enhanced the formation of tubes by human umbilical vein endothelial cells (HUVECs) by approximately 240%, but, when delivered via an AAV vector to the retina of mice, PRELP inhibited laser-induced CNV by 60%. PRELP reduced deposition of MAC in vivo by 25.5%. Our results have implications for the development of complement inhibitors as a therapy for AMD.


Asunto(s)
Neovascularización Coroidal/prevención & control , Proteínas Inactivadoras de Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Proteínas de la Matriz Extracelular/genética , Glicoproteínas/genética , Degeneración Macular/genética , Animales , Ceguera/genética , Coroides/irrigación sanguínea , Coroides/patología , Neovascularización Coroidal/genética , Activación de Complemento , Factor H de Complemento/genética , Proteínas Inactivadoras de Complemento/biosíntesis , Complejo de Ataque a Membrana del Sistema Complemento/biosíntesis , Dependovirus/genética , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/biosíntesis , Terapia Genética , Vectores Genéticos , Glicoproteínas/biosíntesis , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Degeneración Macular/prevención & control , Ratones , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple , Retina/patología , Epitelio Pigmentado de la Retina/patología
18.
Food Funct ; 4(12): 1811-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24150197

RESUMEN

Penthorum chinense Pursh is rich in flavonoids, which have strong antioxidant and anticomplement activities. In order to optimize their extraction conditions, various extraction parameters were chosen to identify their effects on flavonoids extraction. Single factor and Box-Behnken experimental designs consisting of 24 experimental runs and five replicates at zero point were applied to obtain the optimal extraction yield. The optimization conditions for flavonoids extraction were determined as follows: ethanol concentration 60.89%, extraction time 68.15 min, temperature 52.89 °C and liquid/solid ratio 19.70 : 1. The corresponding flavonoids content was 7.19%. The regression equation was found to fit well with the actual situation. Furthermore, the antioxidant activity (the free radical scavenging ability and ferric reducing/antioxidant power) and anticomplement ability of the flavonoids from P. chinense were determined. Results showed that the flavonoids of P. chinense displayed significant antioxidant and anticomplement activities. Its antioxidant activity can compete with ascorbic acid (Vc), whereas its anticomplement activity (IC50 = 111.6 µg ml(-1)) surpassed the effect of heparin (IC50 = 399.7 µg ml(-1)) which was used as the positive control, suggesting that P. chinense flavonoids and their related products could potentially be used as a promising natural agent in the treatment of humoral effector inflammation.


Asunto(s)
Antioxidantes/farmacología , Proteínas Inactivadoras de Complemento/química , Proteínas Inactivadoras de Complemento/farmacología , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Flavonoides/farmacología , Magnoliopsida/química , Animales , Antioxidantes/química , Antioxidantes/aislamiento & purificación , Proteínas Inactivadoras de Complemento/aislamiento & purificación , Medicamentos Herbarios Chinos/aislamiento & purificación , Eritrocitos/efectos de los fármacos , Eritrocitos/fisiología , Flavonoides/química , Flavonoides/aislamiento & purificación , Ovinos
19.
Molecules ; 18(4): 4054-80, 2013 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-23563857

RESUMEN

Protostane triterpenes belong to a group of tetracyclic triterpene that exhibit unique structural characteristics. Their natural distribution is primarily limited to the genus Alisma of the Alismataceae family, but they have also been occasionally found in other plant genera such as Lobelia, Garcinia, and Leucas. To date, there are 59 known protostane structures. Many of them have been reported to possess biological properties such as improving lipotropism, hepatoprotection, anti-viral activity against hepatitis B and HIV-I virus, anti-cancer activity, as well as reversal of multidrug resistance in cancer cells. On the other hand, fusidanes are fungal products characterized by 29-nor protostane structures. They possess antibiotic properties against staphylococci, including the methicillin-resistant Staphylococcus aureus (MRSA). Fusidic acid is a representative member which has found clinical applications. This review covers plant sources of the protostanes, their structure elucidation, characteristic structural and spectral properties, as well as biological activities. The fungal sources, structural features, biological activities of fusidanes are also covered in this review. Additionally, the biogenesis of these two types of triterpenes is discussed and a refined pathway is proposed.


Asunto(s)
Extractos Vegetales/química , Triterpenos/química , Alisma/química , Antibacterianos/química , Antibacterianos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Antivirales/análisis , Antivirales/química , Antivirales/farmacología , Proteínas Inactivadoras de Complemento/química , Proteínas Inactivadoras de Complemento/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Ácido Fusídico/química , Ácido Fusídico/farmacología , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Espectroscopía de Resonancia Magnética , Extractos Vegetales/farmacología , Triterpenos/farmacología
20.
FASEB J ; 27(6): 2355-66, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23482636

RESUMEN

Recently discovered Sushi domain-containing protein 4 (SUSD4) contains several Sushi or complement control protein domains; therefore, we hypothesized that it may act as complement inhibitor. Two isoforms of human SUSD4, fused to the Fc part of human IgG, were recombinantly expressed in Chinese hamster ovary (CHO) cells. The secreted soluble isoform of SUSD4 (SUSD4b) inhibited the classical and lectin complement pathways by 50% at a concentration of 0.5 µM. This effect was due to the fact that 1 µM SUSD4b inhibited the formation of the classical C3 convertase by 90%. The membrane-bound isoform (SUSD4a) inhibited the classical and alternative complement pathways when expressed on the surface of CHO cells but not when expressed as a soluble, truncated protein. In all functional studies, we used known complement inhibitors as positive controls, while Coxsackie adenovirus receptor, which has no effect on complement, expressed with Fc tag, was a negative control. We also studied the mRNA expression of both isoforms of SUSD4 in a panel of human tissues using quantitative PCR and primarily found SUSD4a in esophagus and brain, while SUSD4b was highly expressed in esophagus, ovary, and heart. Overall, our results show that SUSD4 is a novel complement inhibitor with restricted expression.


Asunto(s)
C3 Convertasa de la Vía Clásica del Complemento/antagonistas & inhibidores , C3 Convertasa de la Vía Clásica del Complemento/biosíntesis , Proteínas Inactivadoras de Complemento/fisiología , Proteínas de la Membrana/fisiología , Animales , Células CHO , Complemento C3/metabolismo , Proteínas Inactivadoras de Complemento/genética , Vía Clásica del Complemento , Cricetinae , Cricetulus , Femenino , Humanos , Inmunidad Innata/genética , Proteínas de la Membrana/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA