Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 288
Filtrar
1.
Naunyn Schmiedebergs Arch Pharmacol ; 396(10): 2729-2739, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37126195

RESUMEN

The digestive system is exposed to severe inflammation as a result of taking some medications that have gastrointestinal side effects. Sixty Swiss-albino male mice were randomly distributed into six groups to treat inflammations of the colon, stomach, and small intestine caused by taking high doses of diclofenac (D), with two novel synthesized compounds, pyrazolo [3,4 d] pyridazine derivatives (Co1 and Co2). Myeloperoxidase enzyme activity was determined in the colon and small intestinal tissues. Serum contents of TNF-α, IL-22, IgG, and IgM were determined by ELISA. Histopathological examinations of the colon, small intestinal, and stomach tissues were microscopically analyzed. TNF-α, IL-22, and TNFSF11 gene expression were measured in the colon, intestinal, and spleen using qRT-PCR. Diclofenac caused surface columnar epithelial cell loss, focal necrosis of the gastric mucosa, inflammatory cell infiltration, and congested blood vessels in the stomach, colon, and small intestinal tissues. Co1 component was found to be better than Co2 component in reducing the focal necrosis of gastric mucosa and improving the histological structures of the stomach, colon, and small intestinal tissues. After 14 days, the activity of the myeloperoxidase enzyme was increased in group D and decreased in groups DCo1, DCo2, Co1, and Co2. Serum concentrations of TNF-α and IgG were increased, while IL-22 and IGM were reduced in the D, DCo1, and DCo2 groups compared with the Co1 and control groups. TNF-α gene was upregulated in the D group and downregulated in the Co1 group, while the IL-22 gene was downregulated in the D group and upregulated in the Co1 group compared with the control group. The CO1 component may be useful in reducing digestive system inflammation.


Asunto(s)
Colitis , Ratones , Animales , Colitis/tratamiento farmacológico , Peroxidasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Diclofenaco/farmacología , Dióxido de Carbono/metabolismo , Dióxido de Carbono/farmacología , Dióxido de Carbono/uso terapéutico , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Inflamación/patología , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Colon , Antioxidantes/farmacología , Necrosis/tratamiento farmacológico , Necrosis/metabolismo , Necrosis/patología , Inmunoglobulina G/metabolismo , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Inmunoglobulina M/metabolismo , Inmunoglobulina M/farmacología , Inmunoglobulina M/uso terapéutico , Modelos Animales de Enfermedad
2.
Artículo en Inglés | MEDLINE | ID: mdl-36030005

RESUMEN

Pyraclostrobin (PYR), a strobilurin fungicide, has been widely used to control fungal diseases, posing potential risk to aquatic organisms. However, the toxic effects of PYR to fish remained largely unknown. In this study, common carp (Cyprinus carpio L.) was exposed to environmentally relevant levels of PYR (0, 0.5 and 5.0 µg/L) for 30 days to assess its chronic toxicity and potential toxicity mechanism. The results showed that long-term exposure to PYR induced hepatopancreas damage as evident by increased in serum transaminase activities (AST and ALT). Moreover, PYR exposure remarkably enhanced the expressions of hsp70 and hsp90, decreased the levels of antioxidant enzymes and biomarkers and promoted the reactive oxygen species (H2O2 and O2-) and MDA contents in carp hepatopancreas. PYR exposure also upregulated apoptosis-related genes (bax, apaf-1, caspase-3 and caspase-9) and reduced anti-apoptosis gene bcl-2 in fish hepatopancreas. Moreover, PYR exposure altered the expressions of inflammatory cytokines (IL-1ß, IL-6, TNF-α and TGF-ß) in the serum and hepatopancreas and the level of NF-κB p65 in the hepatopancreas. Further research indicated that PYR exposure markedly changed the levels of immune parameters (LYZ, C3, IgM, ACP and AKP) in the serum and/or hepatopancreas, indicating that chronic PYR exposure also has immunotoxicity on fish. Additionally, we found that PYR exposure upregulated p38 and jnk MAPK transcription levels, suggesting that MAPK may be play important role in PYR-induced apoptosis and inflammatory response in the hepatopancreas of common carp. In summary, PYR exposure induced oxidative stress, triggered apoptosis, inflammatory and immune response in common carp, which can help to elucidate the possible toxicity mechanism of PYR in fish.


Asunto(s)
Carpas , Fungicidas Industriales , Animales , Antioxidantes/metabolismo , Carpas/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Fungicidas Industriales/toxicidad , Hepatopáncreas/metabolismo , Peróxido de Hidrógeno/metabolismo , Inmunoglobulina M/metabolismo , Inmunoglobulina M/farmacología , Interleucina-6/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Estrobilurinas/metabolismo , Estrobilurinas/toxicidad , Transaminasas , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Proteína X Asociada a bcl-2/metabolismo
3.
Sci Total Environ ; 850: 157997, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-35964742

RESUMEN

The purpose of this study was to investigate the effects of dietary aflatoxin B1 (AFB1) on growth performance and AFB1 biotransformation, and hepatic oxidative stress, endoplasmic reticulum (ER) stress, apoptosis, and inflammation in northern snakehead (Channa argus). A total of 600 northern snakeheads (7.52 ± 0.02 g) were divided into five groups (three replicates/group) and fed the diets with AFB1 at concentrations of 0, 50, 100, 200, and 400 ppb for 8 weeks. The results demonstrated that dietary AFB1 (≥ 200 ppb) reduced FBW, WG, and SGR. 100, 200, and 400 ppb AFB1 treatment groups significantly decreased the PER, CRP, C3, C4, IgM, and LYS levels in northern snakehead, while FCR was significant increased. Moreover, dietary AFB1 (100, 200, and 400 ppb) increased cyp1a, cyp1b (except 400 ppb), and cyp3a mRNA expression levels, while reducing the GST enzymatic activity and mRNA expression levels in northern snakehead. Furthermore, AFB1 (≥ 100 ppb) increased ROS, MDA, and 8-OHdG levels, and grp78, ire1, perk, jnk, chop, and traf2 mRNA expression levels, and decreased SOD, CAT, GSH-Px, and GSH (except 100 ppb) levels and the gene expression levels of cat, gsh-px (except 100 ppb), and Cu/Zn sod. In addition, AFB1 (100, 200, and 400 ppb) up-regulated the cyt-c, bax, cas-3, and cas-9 mRNA levels in the liver, while down-regulating the bcl-2 expression levels. Meanwhile, the expression levels of nf-κb, tnf-α (except 100 ppb), il-1ß, and il-8 in the liver were up-regulated in AFB1 treatment groups (≥ 100 ppb), while the iκbα mRNA levels were down-regulated. In summary, dietary AFB1 reduced growth performance and humoral immunity in northern snakehead. Meanwhile, the cyclic occurrence of oxidative stress and ER stress, and induced apoptosis and inflammation, is one of the main reasons for AFB1-induced liver injury in the northern snakehead, which will provide valuable information and a fresh perspective for further research into AFB1-induced liver injury in fish.


Asunto(s)
Aflatoxina B1 , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Aflatoxina B1/toxicidad , Animales , Apoptosis , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Citocromo P-450 CYP3A/metabolismo , Dieta/veterinaria , Estrés del Retículo Endoplásmico , Peces/metabolismo , Inmunoglobulina M/metabolismo , Inmunoglobulina M/farmacología , Inflamación/inducido químicamente , Interleucina-8/metabolismo , Hígado/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor 2 Asociado a Receptor de TNF/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/farmacología
4.
Environ Sci Pollut Res Int ; 29(50): 75344-75355, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35653021

RESUMEN

Arsenic is a toxic heavy metal widely found in the natural environment and has adverse effects on the health of waterfowl and human. Curcumin (CUR), a natural pigment of the golden spice turmeric, exhibits excellent anti-tumor, anti-inflammatory and anti-oxidant activities. But the effects of CUR on duck spleen exposed to arsenic remain largely unknown. In this study, 75 ducks were divided randomly into Control, L-ATO, M-ATO, H-ATO and CUR + H-ATO groups to systematically analyze the underlying role of CUR. The results showed that arsenic trioxide (ATO) led to growth retardation of ducks, hyaline degeneration and sparse cell arrangement on their spleen. And in the ATO-exposed ducks, the levels of immunoglobulins (Ig; IgA, IgG, IgM) in the serum and the expression of autophagy-related genes (Atg5, P62, LC3I, LC3II, LC3II/I, Beclin-1) were significantly upregulated compared with the control ducks. Moreover, ATO also activated NF-κB signal pathway and upregulated the expression of pro-inflammatory cytokines (TNF-α, IFN-γ, IL-1ß, IL-2, IL-18). Meanwhile, application of CUR alleviated the ATO toxicity with the release of growth inhibition, and the reduced hyaline degeneration and distortion of the spleen capsule. CUR also suppressed ATO-induced NF-κB activation, pro-inflammatory cytokine addition and expression of autophagy-related genes. Overall, these results suggested that CUR might exert a protective effect against ATO-induced immunosuppression in ducks via anti-inflammation and autophagy restoring.


Asunto(s)
Arsénico , Curcumina , Metales Pesados , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Arsénico/farmacología , Trióxido de Arsénico , Autofagia , Beclina-1/farmacología , Curcumina/farmacología , Curcumina/uso terapéutico , Citocinas , Patos/metabolismo , Inmunoglobulina A/farmacología , Inmunoglobulina A/uso terapéutico , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Inmunoglobulina M/farmacología , Inmunoglobulina M/uso terapéutico , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Interleucina-18/farmacología , Interleucina-18/uso terapéutico , Interleucina-2/farmacología , Interleucina-2/uso terapéutico , Metales Pesados/farmacología , FN-kappa B/metabolismo , Bazo/metabolismo , Factor de Necrosis Tumoral alfa
5.
Toxicol In Vitro ; 81: 105334, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35182770

RESUMEN

Present investigation was carried out to study toxicological damages of copper exposure and mitigation of its adverse effects with ß-glucan administration in IgM+ B cells which processes multiple roles similar to macrophages in Nile tilapia (Oreochromis niloticus). IgM+ B cells were pretreated with ß-glucan (25 µg/mL) for 24 h before exposed to cupric oxide nanoparticles (CuO NPs) or cupric chloride (Cu ions) at the doses of 0, 5, 10, and 20 µg/mL for 24 h, respectively. Our results demonstrated that ß-glucan increased reduced glutathione (GSH) to against oxidative damage from CuO NPs and Cu ions exposure in IgM+ B cells. The apoptosis process through mitochondrial signaling pathway was depressed in IgM+ B cells since the mitochondrial membrane potential (ΔΨm) was protected from copper exposure by ß-glucan treatment. Furthermore, the inhibition on phagocytic abilities of IgM+ B cells caused by copper exposure could be enhanced with ß-glucan treatment via evaluation of microspheres and bioparticles uptake and LPS-induced NO production. Importantly, ß-glucan might participate in immunomodulation in IgM+ B cells through B cell antigen receptor (BCR) to suppress toxicological effect derived from copper exposure. Taken together, this study provides more information on the toxicological damages in IgM+ B cells upon copper exposure and explains the molecular mechanism to reverse adverse effects caused by copper exposure with ß-glucan administration.


Asunto(s)
Cíclidos , beta-Glucanos , Animales , Cíclidos/metabolismo , Cobre/toxicidad , Inmunoglobulina M/metabolismo , Inmunoglobulina M/farmacología , Estrés Oxidativo , beta-Glucanos/metabolismo , beta-Glucanos/farmacología
6.
Mol Cancer Ther ; 20(12): 2483-2494, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34711645

RESUMEN

Death receptor 5 (DR5) is an attractive target for cancer therapy due to its broad upregulated expression in multiple cancers and ability to directly induce apoptosis. Though anti-DR5 IgG antibodies have been evaluated in clinical trials, limited efficacy has been attributed to insufficient receptor crosslinking. IGM-8444 is an engineered, multivalent agonistic IgM antibody with 10 binding sites to DR5 that induces cancer cell apoptosis through efficient DR5 multimerization. IGM-8444 bound to DR5 with high avidity and was substantially more potent than an IgG with the same binding domains. IGM-8444 induced cytotoxicity in a broad panel of solid and hematologic cancer cell lines but did not kill primary human hepatocytes in vitro, a potential toxicity of DR5 agonists. In multiple xenograft tumor models, IGM-8444 monotherapy inhibited tumor growth, with strong and sustained tumor regression observed in a gastric PDX model. When combined with chemotherapy or the BCL-2 inhibitor ABT-199, IGM-8444 exhibited synergistic in vitro tumor cytotoxicity and enhanced in vivo efficacy, without augmenting in vitro hepatotoxicity. These results support the clinical development of IGM-8444 in solid and hematologic malignancies as a monotherapy and in combination with chemotherapy or BCL-2 inhibition.


Asunto(s)
Antineoplásicos/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Genes bcl-2/genética , Inmunoglobulina M/uso terapéutico , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Sulfonamidas/uso terapéutico , Animales , Antineoplásicos/farmacología , Apoptosis , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoglobulina M/farmacología , Ratones , Ratones Desnudos , Sulfonamidas/farmacología
7.
Front Immunol ; 11: 575154, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178202

RESUMEN

Natural IgM antibodies (NAbs) have been shown to recognize injury-associated neoepitopes and to initiate pathogenic complement activation. The NAb termed C2 binds to a subset of phospholipids displayed on injured cells, and its role(s) in arthritis, as well as the potential therapeutic benefit of a C2 NAb-derived ScFv-containing protein fused to a complement inhibitor, complement receptor-related y (Crry), on joint inflammation are unknown. Our first objective was to functionally test mAb C2 binding to apoptotic cells from the joint and also evaluate its inflammation enhancing capacity in collagen antibody-induced arthritis (CAIA). The second objective was to generate and test the complement inhibitory capacity of C2-Crry fusion protein in the collagen-induced arthritis (CIA) model. The third objective was to demonstrate in vivo targeting of C2-Crry to damaged joints in mice with arthritis. The effect of C2-NAb on CAIA in C57BL/6 mice was examined by inducing a suboptimal disease. The inhibitory effect of C2-Crry in DBA/1J mice with CIA was determined by injecting 2x per week with a single dose of 0.250 mg/mouse. Clinical disease activity (CDA) was examined, and knee joints were fixed for analysis of histopathology, C3 deposition, and macrophage infiltration. In mice with suboptimal CAIA, at day 10 there was a significant (p < 0.017) 74% increase in the CDA in mice treated with C2 NAb, compared to mice treated with F632 control NAb. In mice with CIA, at day 35 there was a significant 39% (p < 0.042) decrease in the CDA in mice treated with C2-Crry. Total scores for histopathology were also 50% decreased (p < 0.0005) in CIA mice treated with C2-Crry. C3 deposition was significantly decreased in the synovium (44%; p < 0.026) and on the surface of cartilage (42%; p < 0.008) in mice treated with C2-Crry compared with PBS treated CIA mice. Furthermore, C2-Crry specifically bound to apoptotic fibroblast-like synoviocytes in vitro, and also localized in the knee joints of arthritic mice as analyzed by in vivo imaging. In summary, NAb C2 enhanced arthritis-related injury, and targeted delivery of C2-Crry to inflamed joints demonstrated disease modifying activity in a mouse model of human inflammatory arthritis.


Asunto(s)
Antirreumáticos/farmacología , Artritis Experimental/tratamiento farmacológico , Activación de Complemento/efectos de los fármacos , Inmunoglobulina M/farmacología , Articulaciones/efectos de los fármacos , Receptores de Complemento 3b/metabolismo , Anticuerpos de Cadena Única/farmacología , Sinoviocitos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Experimental/patología , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Articulaciones/inmunología , Articulaciones/metabolismo , Articulaciones/patología , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Proteínas Recombinantes de Fusión/farmacología , Sinoviocitos/inmunología , Sinoviocitos/metabolismo , Sinoviocitos/patología , Timocitos/efectos de los fármacos , Timocitos/inmunología , Timocitos/metabolismo , Timocitos/patología
8.
Front Immunol ; 11: 2122, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32983170

RESUMEN

Moraxella catarrhalis is a human-specific commensal of the respiratory tract and an opportunistic pathogen. It is one of the leading cause of otitis media in children and of acute exacerbations in patients with chronic obstructive pulmonary disease, resulting in significant morbidity and economic burden. Vaccines and new immunotherapeutic strategies to treat this emerging pathogen are needed. Complement is a key component of innate immunity that mediates the detection, response, and subsequent elimination of invading pathogens. Many pathogens including M. catarrhalis have evolved complement evasion mechanisms, which include the binding of human complement inhibitors such as C4b-binding protein (C4BP) and Factor H (FH). Inhibiting C4BP and FH acquisition by M. catarrhalis may provide a novel therapeutic avenue to treat infections. To achieve this, we created two chimeric proteins that combined the Moraxella-binding domains of C4BP and FH fused to human immunoglobulin Fcs: C4BP domains 1 and 2 and FH domains 6 and 7 fused to IgM and IgG Fc, respectively. As expected, FH6-7/IgG displaced FH from the bacterial surface while simultaneously activating complement via Fc-C1q interactions, together increasing pathogen elimination. C4BP1-2/IgM also increased serum killing of the bacteria through enhanced complement deposition, but did not displace C4BP from the surface of M. catarrhalis. These Fc fusion proteins could act as anti-infective immunotherapies. Many microbes bind the complement inhibitors C4BP and FH through the same domains as M. catarrhalis, therefore these Fc fusion proteins may be promising candidates as adjunctive therapy against many different drug-resistant pathogens.


Asunto(s)
Proteína de Unión al Complemento C4b/farmacología , Factor H de Complemento/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Moraxella catarrhalis/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Animales , Unión Competitiva , Actividad Bactericida de la Sangre , Células CHO , Complemento C3b/análisis , Complemento C3d/análisis , Proteína de Unión al Complemento C4b/genética , Proteína de Unión al Complemento C4b/metabolismo , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Cricetinae , Cricetulus , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Inmunoglobulina G/farmacología , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Inmunoglobulina M/farmacología , Moraxella catarrhalis/metabolismo , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
9.
MAbs ; 12(1): 1818436, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32936727

RESUMEN

Treatment of ocular disease is hindered by the presence of the blood-retinal barrier, which restricts access of systemic drugs to the eye. Intravitreal injections bypass this barrier, delivering high concentrations of drug to the targeted tissue. However, the recommended dosing interval for approved biologics is typically 6-12 weeks, and frequent travel to the physician's office poses a substantial burden for elderly patients with poor vision. Real-world data suggest that many patients are under-treated. Here, we investigate IgMs as a novel platform for treating ocular disease. We show that IgMs are well-suited to ocular administration due to moderate viscosity, long ocular exposure, and rapid systemic clearance. The complement-dependent cytotoxicity of IgMs can be readily removed with a P436G mutation, reducing safety liabilities. Furthermore, dodecavalent binding of IgM hexamers can potently activate pathways implicated in the treatment of progressive blindness, including the Tie2 receptor tyrosine kinase signaling pathway for the treatment of diabetic macular edema, or the death receptor 4 tumor necrosis family receptor pathway for the treatment of wet age-related macular degeneration. Collectively, these data demonstrate the promise of IgMs as therapeutic agonists for treating progressive blindness.


Asunto(s)
Sistemas de Liberación de Medicamentos , Inmunoglobulina M/farmacología , Degeneración Macular , Cuerpo Vítreo/metabolismo , Animales , Células CHO , Cricetulus , Humanos , Inyecciones Intravítreas , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Ratas
10.
Sci Rep ; 9(1): 8097, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31147593

RESUMEN

Described in several epithelial cancer cells, Tn- (GalNAcα1-O-Ser/Thr) and T- (Galß3GalNAcα1-O-Ser/Thr) antigens are examples of tumor-associated antigens. Increased expression of Tn- and T-antigens is associated with tumor invasion and metastasis, and patients with high concentration of anti-Tn and anti-T antibodies have a more benign evolution of pathology. Asialofetuin (ASF) and ovine submaxillary mucin (OSM) are two glycoproteins that expose T- and Tn-antigen, respectively. In this work, using ASF or OSM we affinity-purified anti-T and anti-Tn antibodies from normal human plasma and tested their ability to specifically recognize tumor human tissues. Whereas purified anti-T antibodies (purity degree increase of 127-fold, and 22% recovery) were mainly IgG, for purified anti-Tn antibodies (purity degree enhancement of 125-fold, and 26% yield) the IgM fraction was predominant over the IgG one. IgG2 subclass was significantly enriched in both purified antibody samples. Purified antibodies did not bind normal human tissue (0/42), although recognized malignant tissues from different origin such as colon carcinoma (11/77 by anti-Tn; 7/79 by anti-T), breast carcinoma (10/23 by anti-Tn; 7/23 by anti-T), and kidney carcinoma (45/51 by anti-Tn; 42/51 by anti-T). Our results suggest that purified human anti-Tn and anti-T antibodies have a potential as anti-tumor therapeutic agents; restoring their levels in human sera could positively affect the evolution of patients with epithelial tumor pathologies.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/inmunología , Antineoplásicos Inmunológicos/farmacología , Carcinoma/tratamiento farmacológico , Inmunoglobulina G/farmacología , Inmunoglobulina M/farmacología , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/uso terapéutico , Asialoglicoproteínas/inmunología , Carcinoma/inmunología , Carcinoma/patología , Línea Celular Tumoral , Cromatografía de Afinidad/métodos , Ensayos de Selección de Medicamentos Antitumorales , Fetuínas/inmunología , Humanos , Proteínas Inmovilizadas/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/aislamiento & purificación , Inmunoglobulina G/uso terapéutico , Inmunoglobulina M/inmunología , Inmunoglobulina M/aislamiento & purificación , Inmunoglobulina M/uso terapéutico , Mucinas/inmunología , Plasma/inmunología
11.
J Clin Neuromuscul Dis ; 20(3): 103-110, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30801480

RESUMEN

OBJECTIVE: To assess the intraepidermal nerve fiber density in patients diagnosed with fibromyalgia (FM) and to evaluate the role of IgM binding to trisulfated heparin disaccharide (TS-HDS) in these patients. METHODS: FM is a poorly understood pain disorder with several proposed pathophysiologic mechanisms. It is characterized by widespread pain, fatigue, and sleep abnormalities. Small fiber neuropathy (SFN) has been proposed as an underlying mechanism, and patients with FM have been shown to have a reduction in the intraepidermal nerve fiber density. An underlying inflammatory process that could be a result of autoimmune phenomena has also been suggested. Non-length-dependent SFN (NLDSFN) has been shown to have a higher incidence of autoimmune disease. Twenty-two patients with established diagnosis of FM underwent skin biopsy at 2 sites; 10 cm above the lateral malleolus and 10 cm above the patella. Serum IgM binding to TS-HDS was assayed using an ELISA method. RESULTS: A total of 5/22 patients had positive TS-HDS antibodies; of these, 4 had NLDSFN (P = 0.0393). Comparison with a control group at Washington University showed no significant difference in percentage with TS-HDS antibodies (P = 0.41). When compared with Washington University database of skin biopsy, there was a trend for an increased percentage of NLDSFN in patients with FM (P = 0.06). CONCLUSIONS: This study further supports the hypothesis that a subgroup of patients with FM has SFN. We suggest a correlation between the presence of NLDSFN and TS-HDS antibodies.


Asunto(s)
Disacáridos/metabolismo , Fibromialgia/complicaciones , Fibromialgia/epidemiología , Heparina/análogos & derivados , Inmunoglobulina M/farmacología , Neuropatía de Fibras Pequeñas/epidemiología , Neuropatía de Fibras Pequeñas/etiología , Adulto , Anticuerpos Monoclonales/sangre , Disacáridos/inmunología , Femenino , Heparina/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , Unión Proteica
12.
Methods Mol Biol ; 1904: 53-81, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30539466

RESUMEN

Antibodies are vital components of the adaptive immune system for the recognition and response to foreign antigens. However, some antibodies recognize self-antigens in healthy individuals. These autoreactive antibodies may modulate innate immune functions. IgM natural autoantibodies (IgM-NAAs) are a class of primarily polyreactive immunoglobulins encoded by germline V-gene segments which exhibit low affinity but broad specificity to both foreign and self-antigens. Historically, these autoantibodies were closely associated with autoimmune disease. Nevertheless, not all human autoantibodies are pathogenic and compelling evidence indicates that IgM-NAAs may exert a spectrum of effects from injurious to protective depending upon cellular and molecular context. In this chapter, we review the current state of knowledge regarding the potential physiological and therapeutic roles of IgM-NAAs in different disease conditions such as atherosclerosis, cancer, and autoimmune disease. We also describe the discovery of two reparative IgM-NAAs by our laboratory and delineate their proposed mechanisms of action in central nervous system (CNS) disease.


Asunto(s)
Autoanticuerpos/inmunología , Autoanticuerpos/uso terapéutico , Inmunoglobulina M/inmunología , Inmunoglobulina M/uso terapéutico , Animales , Afinidad de Anticuerpos/inmunología , Formación de Anticuerpos , Especificidad de Anticuerpos/inmunología , Antígenos/inmunología , Autoanticuerpos/metabolismo , Autoanticuerpos/farmacología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Humanos , Inmunoglobulina M/metabolismo , Inmunoglobulina M/farmacología , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/inmunología , Oligodendroglía/metabolismo , Unión Proteica/inmunología , Resultado del Tratamiento
13.
Clin Immunol ; 188: 45-51, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29274388

RESUMEN

OBJECTIVES: B-cells play a crucial role in the pathogenesis of lupus nephritis. Recently, a separate subset has been discovered characterized by expression of Granzyme B. The aim of this study is to investigate this subset in patients with systemic lupus erythematosus (SLE). METHODS: Isolated PBMCs of SLE-patients (n=30) and healthy controls (n=21) were in vitro stimulated with CPG, IgG+IgM and IL-21. Patients were sub-grouped in patients with and without biopsy proven lupus nephritis. B-cells were analyzed for intracellular Granzyme B expression by flow cytometry. RESULTS: The strongest stimulus for Granzyme B secretion of B-cells was IgG+IgM in presence of IL-21. SLE-patients had a significant decreased percentage of Granzyme B+ B-cells in particular SLE-patients with active disease and with lupus nephritis. CONCLUSIONS: The frequency of GrB+ producing B-cells is reduced in SLE patients. This may contribute to an imbalanced B-cell regulation towards effector B-cells which might promote the development of lupus nephritis.


Asunto(s)
Linfocitos B/efectos de los fármacos , Granzimas/inmunología , Interleucinas/farmacología , Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/inmunología , Adulto , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Cultivadas , Femenino , Granzimas/metabolismo , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Inmunoglobulina M/inmunología , Inmunoglobulina M/farmacología , Interleucinas/inmunología , Lupus Eritematoso Sistémico/metabolismo , Nefritis Lúpica/metabolismo , Masculino , Persona de Mediana Edad , Receptores de Interleucina-21/inmunología , Receptores de Interleucina-21/metabolismo
14.
Microbiol Immunol ; 61(10): 433-441, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28881485

RESUMEN

Heterologous secondary infections are at increased risk of developing dengue hemorrhagic fever (DHF) because of antibody-dependent enhancement (ADE). IgG subclasses can fix and activate complement and bind to Fcɣ receptors. These factors may also play an important role in the development of ADE and thus in the pathogenesis of DHF. The aim of this study was to analyze the indices of anti-dengue IgG subclasses in adult patients with febrile and hemorrhagic dengue in the acute phase. In 2013, 129 patients with dengue fever (DF) and 57 with DHF in Veracruz, Mexico were recruited for this study and anti-dengue IgM and IgG determined by capture ELISA. Anti-dengue IgG subclasses were detected by indirect ELISA. Anti-dengue IgG2 and IgG3 subclasses were detected in patients with dengue. IgG1 increased significantly in the sera of patients with both primary and secondary infections and DHF, but was higher in patients with secondary infections. The IgG4 subclass index was significantly higher in the sera of patients with DHF than in that of those with DF, who were in the early and late acute phase of both primary and secondary infection. In conclusion, indices of subclasses IgG1 and IgG4 were higher in patients with DHF.


Asunto(s)
Anticuerpos Antivirales/sangre , Virus del Dengue/inmunología , Dengue/inmunología , Inmunoglobulina G/sangre , Dengue Grave/inmunología , Adulto , Dengue/virología , Virus del Dengue/clasificación , Virus del Dengue/aislamiento & purificación , Virus del Dengue/patogenicidad , Femenino , Humanos , Inmunoglobulina G/clasificación , Inmunoglobulina M/sangre , Inmunoglobulina M/farmacología , Linfocitos/virología , Masculino , México , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/virología , Neutrófilos/inmunología , Neutrófilos/virología , ARN Viral/análisis , Serotipificación , Dengue Grave/virología , Índice de Severidad de la Enfermedad , Adulto Joven
15.
Neurobiol Dis ; 105: 142-155, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28576706

RESUMEN

Failure of oligodendrocyte precursor cells (OPCs) to differentiate and remyelinate axons is thought to be a major cause of the limited ability of the central nervous system to repair plaques of immune-mediated demyelination in multiple sclerosis (MS). Current therapies for MS aim to lessen the immune response in order to reduce the frequency and severity of attacks, but these existing therapies do not target remyelination or stimulate repair of the damaged tissue. Thus, the promotion of OPC differentiation and remyelination is potentially an important therapeutic goal. Previous studies have shown that a recombinant human-derived monoclonal IgM antibody, designated rHIgM22, promotes remyelination, particularly of the spinal cord in rodent models of demyelination. Here, we examined the effects of rHIgM22 in remyelination in the brain using the mouse model of cuprizone-induced demyelination, which is characterized by spontaneous remyelination. The myelination state of the corpus callosum of cuprizone-fed mice treated with rHIgM22 was examined immediately after the end of the cuprizone diet as well as at different time points during the recovery period with regular food, and compared with that of cuprizone-fed animals treated with either vehicle or human IgM isotype control antibody. Mice fed only regular food were used as controls. We demonstrate that treatment with rHIgM22 accelerated remyelination of the demyelinated corpus callosum. The remyelination-enhancing effects of rHIgM22 were found across different, anatomically distinct regions of the corpus callosum, and followed a spatiotemporal pattern that was similar to that of the spontaneous remyelination process. These enhancing effects were also accompanied by increased differentiation of OPCs into mature oligodendrocytes. Our data indicate strong remyelination-promoting capabilities of rHIgM22 and further support its therapeutic potential in MS.


Asunto(s)
Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Inmunoglobulina M/uso terapéutico , Inhibidores de la Monoaminooxidasa/toxicidad , Remielinización/efectos de los fármacos , Análisis de Varianza , Animales , Diferenciación Celular/efectos de los fármacos , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Enfermedades Desmielinizantes/tratamiento farmacológico , Modelos Animales de Enfermedad , Inmunoglobulina M/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/inmunología , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/patología , Nervio Óptico/efectos de los fármacos , Nervio Óptico/metabolismo , Nervio Óptico/patología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Tiempo
16.
Hum Antibodies ; 25(3-4): 121-129, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28269761

RESUMEN

A single peripheral dose of CNS-binding IgMs promote remyelination and preserve axons in a number of animal models of neurologic disease. A myelin-binding recombinant human IgM (rHIgM22) is presently in a safety trial in MS patients following an acute MS exacerbation. rHIgM22 (directed against oligodendrocytes) or rHIgM12 (directed against neurons) were administered to mice with MOG-induced experimental autoimmune encephalomyelitis (EAE) with study endpoints: clinical deficits and brain and spinal cord pathology. IgMs were administered at a therapeutic dose of 100 µ g intra peritoneal at the time of immunization (day -1, 0, +$1), disease onset (15 days) or peak of the disease (28 days). Disease course was not worsened by either human IgM regardless of the time of treatment. Of note, the human IgM that recognizes a carbohydrate epitope on gangliosides and NCAM, rHIgM12, reduced brain pathology when given at time of immunization or at onset of disease, but did not reduce clinical deficits or spinal cord disease burden. Hence, treatment with rHIgM12 resulted in marked reduction in meningeal inflammation. Data consistent with the hypothesis that in the EAE model this molecule has an immune-modulatory effect. Treatment with an anti-CD4 blocking IgG prevented both clinical course and CNS pathology. This pre-clinical study further supports the safety of therapeutic CNS-binding human IgMs in the presence of autoimmunity and clearly differentiates them from IgGs directed against MOG or aquaporin-4 that worsen neurologic disease.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Enfermedades Desmielinizantes/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunoglobulina M/farmacología , Factores Inmunológicos/farmacología , Molécula L1 de Adhesión de Célula Nerviosa/inmunología , Fármacos Neuroprotectores/farmacología , Ácidos Siálicos/inmunología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/inmunología , Disfunción Cognitiva/patología , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Esquema de Medicación , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Adyuvante de Freund/administración & dosificación , Humanos , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/inmunología , Vaina de Mielina/patología , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/inmunología , Oligodendroglía/patología , Fragmentos de Péptidos/administración & dosificación , Unión Proteica , Proteínas Recombinantes/farmacología , Ácidos Siálicos/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/inmunología , Médula Espinal/patología
17.
Sci Rep ; 7: 42989, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28230186

RESUMEN

IgM is the first antibody to be produced in immune responses and plays an important role in the neutralization of bacteria and viruses. Human IgM is heavily glycosylated, featuring five N-linked glycan sites on the µ chain and one on the J-chain. Glycosylation of IgG is known to modulate the effector functions of Fcγ receptors. In contrast, little is known about the effect of glycosylation on IgM binding to the human Fcµ receptor (hFCMR). In this study, we identify the Cµ4 domain of IgM as the target of hFCMR, and show that binding and internalization of IgM by hFCMR is glycan-independent. We generated a homology-based structure for hFCMR and used molecular dynamic simulations to show how this interaction with IgM may occur. Finally, we reveal an inhibitory function for IgM in the proliferation of T cells.


Asunto(s)
Inmunoglobulina M/metabolismo , Polisacáridos/química , Receptores Fc/metabolismo , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Endocitosis , Glicosilación , Humanos , Inmunoglobulina M/química , Inmunoglobulina M/farmacología , Simulación de Dinámica Molecular , Fitohemaglutininas/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Receptores Fc/química , Linfocitos T/citología , Linfocitos T/metabolismo
18.
Int Immunopharmacol ; 40: 11-15, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27567246

RESUMEN

Intravenous immunoglobulin (IVIG), consisting of IgG, is the first-line treatment for Guillain-Barré syndrome and multifocal motor neuropathy. IgG, but neither IgM nor IgA, has been demonstrated in vitro to inhibit complement deposition mediated by anti-ganglioside autoantibodies in sera from patients with both conditions. The objective of this study is to investigate the in vitro effectiveness of IgM and IgA in inhibiting complement deposition to ganglioside/anti-ganglioside antibody complexes. Serum samples were obtained from patients with multifocal motor neuropathy associated with anti-GM1 IgM antibodies, Guillain-Barré syndrome associated with anti-GM1 IgG antibodies and Miller Fisher syndrome associated with anti-GQ1b IgG antibodies. Inhibition of complement deposition using different immunoglobulin preparations was measured by enzyme-linked immunosorbent assay. IgM/A-enriched IVIG and immunoglobulin isotypes (polyclonal IgM and IgA) showed higher potential in inhibiting complement deposition than standard IVIG. Although the safety concerns about the use of IgM and IgA for an immunotherapy still remain, IgM and IgA may serve as an alternative immunotherapy in those anti-ganglioside antibody-mediated neuropathies.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Síndrome de Guillain-Barré/inmunología , Inmunoglobulina A/farmacología , Inmunoglobulina M/farmacología , Inmunoglobulinas Intravenosas/farmacología , Anticuerpos Antiidiotipos/sangre , Anticuerpos Antiidiotipos/inmunología , Síndrome de Guillain-Barré/sangre , Humanos
19.
PLoS One ; 11(8): e0160907, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27504630

RESUMEN

INTRODUCTION: Sepsis remains associated with a high mortality rate. Endotoxin has been shown to influence viscoelastic coagulation parameters, thus suggesting a link between endotoxin levels and the altered coagulation phenotype in septic patients. This study evaluated the effects of systemic polyspecific IgM-enriched immunoglobulin (IgM-IVIg) (Pentaglobin® [Biotest, Dreieich, Germany]) on endotoxin activity (EA), inflammatory markers, viscoelastic and conventional coagulation parameters. METHODS: Patients with severe sepsis were identified by daily screening in a tertiary, academic, surgical ICU. After the inclusion of 15 patients, the application of IgM-IVIg (5 mg/kg/d over three days) was integrated into the unit's standard operation procedure (SOP) to treat patients with severe sepsis, thereby generating "control" and "IgM-IVIg" groups. EA assays, thrombelastometry (ROTEM®) and impedance aggregometry (Multiplate®) were performed on whole blood. Furthermore, routine laboratory parameters were determined according to unit's standards. RESULTS: Data from 26 patients were included. On day 1, EA was significantly decreased in the IgM-IVIg group following 6 and 12 hours of treatment (0.51 ±0.06 vs. 0.26 ±0.07, p<0.05 and 0.51 ±0.06 vs. 0.25 ±0.04, p<0.05) and differed significantly compared with the control group following 6 hours of treatment (0.26 ±0.07 vs. 0.43 ±0.07, p<0.05). The platelet count was significantly higher in the IgM-IVIg group following four days of IgM-IVIg treatment (200/nl ±43 vs. 87/nl ±20, p<0.05). The fibrinogen concentration was significantly lower in the control group on day 2 (311 mg/dl ±37 vs. 475 mg/dl ±47 (p = 0.015)) and day 4 (307 mg/dl ±35 vs. 420 mg/dl ±16 (p = 0.017)). No differences in thrombelastometric or aggregometric measurements, or inflammatory markers (interleukin-6 (IL-6), leukocyte, lipopolysaccharide binding protein (LBP)) were observed. CONCLUSION: Treatment with IgM-enriched immunoglobulin attenuates the EA levels in patients with severe sepsis and might have an effect on septic thrombocytopenia and fibrinogen depletion. Viscoelastic, aggregometric or inflammatory parameters were not influenced. TRIAL REGISTRATION: clinicaltrials.gov NCT02444871.


Asunto(s)
Endotoxinas/toxicidad , Inmunoglobulina M/farmacología , Sepsis/inducido químicamente , Sepsis/tratamiento farmacológico , Anciano , Biomarcadores/metabolismo , Estudios de Cohortes , Femenino , Humanos , Inmunoglobulina M/uso terapéutico , Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Sepsis/metabolismo
20.
Arthritis Rheumatol ; 68(12): 2917-2928, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27274010

RESUMEN

OBJECTIVE: In vitro studies suggest that the type I interferon (IFN) signature seen in most lupus patients results from Fcγ receptor-mediated uptake of nucleic acid-containing immune complexes by plasmacytoid dendritic cells and engagement of endosomal Toll-like receptors. The aim of this study was to reexamine the pathogenesis of the IFN signature in vivo. METHODS: Lupus was induced in mice by injecting pristane. Some mice were treated with normal immunoglobulin or with cobra venom factor to deplete complement. The IFN signature was evaluated by polymerase chain reaction. The IFN signature also was determined in C4-deficient patients and control subjects. RESULTS: Wild-type C57BL/6 mice with pristane-induced lupus developed a strong IFN signature, which was absent in immunoglobulin-deficient (µMT), C3-/- , and CD18-/- mice. Intravenous infusion of normal IgM, but not IgG, restored the IFN signature in µMT mice, and the IFN signature in wild-type mice was inhibited by depleting complement, suggesting that opsonization by IgM and complement is involved in IFN production. Consistent with that possibility, the levels of "natural" IgM antibodies reactive with dead cells were increased in pristane-treated wild-type mice compared with untreated controls, and in vivo phagocytosis of dead cells was impaired in C3-deficient mice. To examine the clinical relevance of these findings, we identified 10 C4-deficient patients with lupus-like disease and compared them with 152 C4-intact patients and 21 healthy controls. In comparison with C4-intact patients, C4-deficient patients had a different clinical/serologic phenotype and lacked the IFN signature. CONCLUSION: These studies define previously unrecognized roles of natural IgM, complement, and complement receptors in generating the IFN signature in lupus.


Asunto(s)
Complemento C3/genética , Inmunoglobulina M/inmunología , Interferón Tipo I/genética , Lupus Eritematoso Sistémico/genética , Proteínas Opsoninas/inmunología , Adulto , Animales , Apoptosis/inmunología , Autoanticuerpos/inmunología , Antígenos CD18/genética , Estudios de Casos y Controles , Complemento C3/efectos de los fármacos , Complemento C3/inmunología , Complemento C4/deficiencia , Inactivadores del Complemento/farmacología , Proteínas del Sistema Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Venenos Elapídicos/farmacología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Inmunoglobulina G/farmacología , Inmunoglobulina M/farmacología , Inmunosupresores/toxicidad , Interferón Tipo I/efectos de los fármacos , Interferón Tipo I/inmunología , Lupus Eritematoso Sistémico/inmunología , Masculino , Persona de Mediana Edad , Fagocitosis/inmunología , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Terpenos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...