Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Carbohydr Polym ; 250: 116869, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33049818

ABSTRACT

Melanoma is the most lethal form of skin cancer, with a worldwide increase in incidence. Despite the increased overall survival of metastatic melanoma patients given recent advances in targeted and immunotherapy, it still has a poor prognosis and available treatment options carry diverse severe side effects. Polysaccharides from seaweed have been shown to exert antitumor activities. Here we show in vitro and in vivo antitumor activities of a sulfated homogalactan (named 3G4S) from Codium isthmocladum seaweed in the B16-F10 murine melanoma cell line. 3G4S did not induce cytotoxicity or proliferation changes; however, it was able to reduce solid tumor growth and metastasis, while not inducing side effects in mice. B16-F10 cells traits related to the metastatic cascade were also impaired by 3G4S, reducing cell invasion, colony-forming capacity and membrane glycoconjugates. Therefore, 3G4S shows promising antitumor activities without the commonly associated drawbacks of cancer treatments and can be further explored.


Subject(s)
Galactans/pharmacology , Green Chemistry Technology , Melanoma, Experimental/prevention & control , Seaweed/chemistry , Sulfates/chemistry , Animals , Apoptosis , Cell Proliferation , Female , Humans , Male , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured
2.
Sci Rep ; 9(1): 14620, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601823

ABSTRACT

Galectin-3 (Gal-3) controls intercellular and cell-extracellular matrix interactions during immunological responses. In chronic inflammation, Gal-3 is associated with fibrotic events, regulates B cell differentiation and delays lupus progression. Gal-3 deficient mice (Lgals3-/-) have intense germinal center formation and atypical plasma cell generation correlated to high levels IgG, IgE, and IgA. Here, we used pristane (2,6,10,14-tetramethylpentadecane) to induce lupus-like syndrome in Lgals3-/- and Lgals3+/+ BALB/c mice. Mesentery and peritoneal cells were monitored because promptly react to pristane injected in the peritoneal cavity. For the first time, mesenteric tissues have been associated to the pathogenesis of experimental lupus-like syndrome. In Lgals3+/+ pristane-induced mice, mesentery was hallmarked by intense fibrogranulomatous reaction restricted to submesothelial regions and organized niches containing macrophages and B lymphocytes and plasma cells. In contrast, Lgals3-/- pristane-treated mice had diffuse mesenteric fibrosis affecting submesothelium and peripheral tissues, atypical M1/M2 macrophage polarization and significant DLL1+ cells expansion, suggesting possible involvement of Notch/Delta pathways in the disease. Early inflammatory reaction to pristane was characterized by significant disturbances on monocyte recruitment, macrophage differentiation and dendritic cell (DC) responses in the peritoneal cavity of pristane-induced Lgals3-/- mice. A correlative analysis showed that mesenteric damages in the absence of Gal-3 were directly associated with severe portal inflammation and hepatitis. In conclusion, it has suggested that Gal-3 orchestrates histological organization in the mesentery and prevents lupoid hepatitis in experimental lupus-like syndrome by controlling macrophage polarization, Notch signaling pathways and DC differentiation in mesenteric structures.


Subject(s)
Galectin 3/metabolism , Hepatitis/immunology , Lupus Erythematosus, Systemic/immunology , Macrophages, Peritoneal/immunology , Mesentery/pathology , Animals , Disease Models, Animal , Female , Fibrosis , Galectin 3/genetics , Hepatitis/pathology , Humans , Injections, Intraperitoneal , Liver/immunology , Liver/pathology , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/complications , Mesentery/cytology , Mesentery/immunology , Mice , Mice, Knockout , Terpenes/administration & dosage , Terpenes/immunology
3.
Braz J Med Biol Res ; 51(3): e7214, 2018 Jan 11.
Article in English | MEDLINE | ID: mdl-29340530

ABSTRACT

A biosimilar is a biologic product that is similar to a reference biopharmaceutical product, the manufacturing process of which hinders the ability to identically replicate the structure of the original product, and therefore, it cannot be described as an absolute equivalent of the original medication. The currently available technology does not allow for an accurate copy of complex molecules, but it does allow the replication of similar molecules with the same activity. As biosimilars are about to be introduced in oncology practice, these must be evaluated through evidence-based medicine. This manuscript is a position paper, where the Brazilian Society of Clinical Oncology (SBOC) aims to describe pertinent issues regarding the approval and use of biosimilars in oncology. As a working group on behalf of SBOC, we discuss aspects related to definition, labeling/nomenclature, extrapolation, interchangeability, switching, automatic substitution, clinical standards on safety and efficacy, and the potential impact on financial burden in healthcare. We take a stand in favor of the introduction of biosimilars, as they offer a viable, safe, and cost-effective alternative to the biopharmaceutical products currently used in cancer. We hope this document can provide valuable information to support therapeutic decisions that maximize the clinical benefit for the thousands of cancer patients in Brazil and can contribute to expedite the introduction of this new drug class in clinical practice. We expect the conveyed information to serve as a basis for further discussion in Latin America, this being the first position paper issued by a Latin American Oncology Society.


Subject(s)
Biosimilar Pharmaceuticals/therapeutic use , Medical Oncology , Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Biosimilar Pharmaceuticals/economics , Biosimilar Pharmaceuticals/standards , Brazil , Clinical Trials as Topic , Evidence-Based Medicine , Humans , Neoplasms/immunology , Pharmacovigilance , Societies, Medical
4.
Braz. j. med. biol. res ; 51(3): 7214, 2018. tab
Article in English | LILACS | ID: biblio-889052

ABSTRACT

A biosimilar is a biologic product that is similar to a reference biopharmaceutical product, the manufacturing process of which hinders the ability to identically replicate the structure of the original product, and therefore, it cannot be described as an absolute equivalent of the original medication. The currently available technology does not allow for an accurate copy of complex molecules, but it does allow the replication of similar molecules with the same activity. As biosimilars are about to be introduced in oncology practice, these must be evaluated through evidence-based medicine. This manuscript is a position paper, where the Brazilian Society of Clinical Oncology (SBOC) aims to describe pertinent issues regarding the approval and use of biosimilars in oncology. As a working group on behalf of SBOC, we discuss aspects related to definition, labeling/nomenclature, extrapolation, interchangeability, switching, automatic substitution, clinical standards on safety and efficacy, and the potential impact on financial burden in healthcare. We take a stand in favor of the introduction of biosimilars, as they offer a viable, safe, and cost-effective alternative to the biopharmaceutical products currently used in cancer. We hope this document can provide valuable information to support therapeutic decisions that maximize the clinical benefit for the thousands of cancer patients in Brazil and can contribute to expedite the introduction of this new drug class in clinical practice. We expect the conveyed information to serve as a basis for further discussion in Latin America, this being the first position paper issued by a Latin American Oncology Society.


Subject(s)
Humans , Biosimilar Pharmaceuticals/therapeutic use , Medical Oncology , Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Biosimilar Pharmaceuticals/economics , Biosimilar Pharmaceuticals/standards , Brazil , Clinical Trials as Topic , Evidence-Based Medicine , Neoplasms/immunology , Pharmacovigilance , Societies, Medical
5.
Carbohydr Polym ; 178: 95-104, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29050620

ABSTRACT

A heteropolysaccharide was isolated by cold aqueous extraction from edible mushroom Pleurotus eryngii ("King Oyster") basidiocarps and its biological properties were evaluated. Structural assignments were carried out using mono- and bidimensional NMR spectroscopy, monosaccharide composition, and methylation analyses. A mannogalactan having a main chain of (1→6)-linked α-d-galactopyranosyl and 3-O-methyl-α-d-galactopyranosyl residues, both partially substituted at OH-2 by ß-d-Manp (MG-Pe) single-unit was found. Biological effects of mannogalactan from P. eryngii (MG-Pe) were tested against murine melanoma cells. MG-Pe was non-cytotoxic, but reduced in vitro melanoma cells invasion. Also, 50mg/kg MG-Pe administration to melanoma-bearing C57BL/6 mice up to 10days decreased in 60% the tumor volume compared to control. Additionally, no changes were observed when biochemical profile, complete blood cells count (CBC), organs, and body weight were analyzed. Mg-Pe was shown to be a promising anti-melanoma molecule capable of switching melanoma cells to a non-invasive phenotype with no toxicity to melanoma-bearing mice.


Subject(s)
Fungal Polysaccharides/pharmacology , Galactans/pharmacology , Melanoma/drug therapy , Pleurotus/chemistry , Animals , Fruiting Bodies, Fungal/chemistry , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL
6.
Braz J Med Biol Res ; 50(1): e6153, 2017 Feb 06.
Article in English | MEDLINE | ID: mdl-28177038

ABSTRACT

The authors pay homage to the three founders of the Brazilian Journal of Medical and Biological Research Profs. Lewis Joel Greene, Sérgio Henrique Ferreira and Eduardo Moacyr Krieger for their vision and commitment to divulge the scientific production of developing countries.


Subject(s)
Biomedical Research/history , Periodicals as Topic/history , Brazil , History, 20th Century , History, 21st Century , Humans
7.
Oncogenesis ; 6(1): e296, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28134937

ABSTRACT

A major drawback of radiotherapy is the accelerated growth of the surviving tumor cells. Radiotherapy generates a variety of lipids that bind to the receptor for platelet-activating factor, expressed by cells in the tumor microenvironment. In the present study, using the TC-1 tumor cell line, we found that irradiation induced a twofold increase in receptor expression and generated agonists of receptor. Irradiated cells induced a 20-fold increase in live TC-1 proliferation in vitro. Furthermore, subcutaneous co-injection of irradiated TC-1 cells with TC-1 expressing luciferase (TC-1 fluc+) markedly increased TC-1 fluc+ proliferation in a receptor-dependent way. Moreover we used a human carcinoma cell line not expressing the PAF receptor (KBM) and the same cell transfected with the receptor gene (KBP). Following co-injection of live KBP cells with irradiated KBM in RAG mice, the tumor growth was significantly increased compared with tumor formed following co-injection of live KBM with irradiated KBM. This tumor cell repopulation correlated with increased infiltration of tumor-promoting macrophages (CD206+). We propose that receptor represents a possible target for improving the efficacy of radiotherapy through inhibition of tumor repopulation.

8.
Braz. j. med. biol. res ; 50(1): e6153, 2017. graf
Article in English | LILACS | ID: biblio-839235

ABSTRACT

The authors pay homage to the three founders of the Brazilian Journal of Medical and Biological Research Profs. Lewis Joel Greene, Sérgio Henrique Ferreira and Eduardo Moacyr Krieger for their vision and commitment to divulge the scientific production of developing countries.


Subject(s)
History, 20th Century , History, 21st Century , Biomedical Research/history , Periodicals as Topic/history , Brazil
9.
Cancer Lett ; 371(2): p. 151-160, 2016.
Article | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib14031

ABSTRACT

Compelling evidence suggests that fibroblast growth factor 2 (FGF2), overexpressed in melanomas, plays an important role in tumor growth, angiogenesis and metastasis. In this study, we evaluated the therapeutic use of a new anti-FGF2 monoclonal antibody (mAb), 3F12E7, using for that the B16-F10 melanoma model. The FGF2 neutralizing effect of this antibody was certified by in vitro assays, which allowed the further track of its possible in vivo application. 3F12E7 mAb could be retained in B16-F10 tumors, as shown by antibody low-pH elution and nuclear medicine studies, and also led to reduction in number and size of metastatic foci in lungs, when treatment starts one day after intravenous injection of B16-F10 cells. Such data were accompanied by decreased CD34(+) tumor vascular density and impaired subcutaneous tumor outgrowth. Treatments starting one week after melanoma cell intravenous injection did not reduce tumor burden, remaining the therapeutic effectiveness restricted to early-adopted regimens. Altogether, the presented anti-FGF2 3F12E7 mAb stands as a promising agent to treat metastatic melanoma tumors in adjuvant settings. (C) 2015 Elsevier Ireland Ltd. All rights reserved.


Subject(s)
Medical Oncology , Allergy and Immunology
10.
Mol. Cell. Biochem ; 415(1-2): p. 119-131, 2016.
Article | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib13791

ABSTRACT

During the last two decades, new insights into proteasome function and its role in several human diseases made it a potential therapeutic target. In this context, Amblyomin-X is a Kunitz-type FXa inhibitor similar to endogenous tissue factor pathway inhibitor (TFPI) and is a novel proteasome inhibitor. Herein, we have demonstrated Amblyomin-X cytotoxicity to different tumor cells lines such as pancreatic (Panc1, AsPC1BxPC3) and melanoma (SK-MEL-5 and SK-MEL-28). Of note, Amblyomin-X was not cytotoxic to normal human fibroblast cells. In addition, Amblyomin-X promoted accumulation of ER stress markers (GRP78 and GADD153) in sensitive (SK-MEL-28) and bortezomib-resistant (Mia-PaCa-2) tumor cells. The intracellular calcium concentration [Ca2+] (i) was slightly modulated in human tumor cells (SK-MEL-28 and Mia-PaCa-2) after 24 h of Amblyomin-X treatment. Furthermore, Amblyomin-X induced mitochondrial dysfunction, cytochrome-c release, PARP cleavage, and activation of caspase cascade in both human tumor (SK-MEL-28 and Mia-PaCa-2) cells. These investigations might help in further understanding of the antitumor properties of Amblyomin-X


Subject(s)
Biochemistry , Medical Oncology , Cell Biology
11.
Oncogene ; 34(10): 1270-9, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-24662834

ABSTRACT

Intratumoral heterogeneity (ITH) represents an obstacle for cancer diagnosis and treatment, but little is known about its functional role in cancer progression. The A Desintegrin And Metalloproteinase 23 (ADAM23) gene is epigenetically silenced in different types of tumors, and silencing is often associated with advanced disease and metastasis. Here, we show that invasive breast tumors exhibit significant ADAM23-ITH and that this heterogeneity is critical for tumor growth and metastasis. We demonstrate that while loss of ADAM23 expression enhances invasion, it causes a severe proliferative deficiency and is not itself sufficient to trigger metastasis. Rather, we observed that, in ADAM23-heterotypic environments, ADAM23-negative cells promote tumor growth and metastasis by enhancing the proliferation and invasion of adjacent A23-positive cells through the production of LGI4 (Leucine-rich Glioma Inactivated 4) and nitric oxide (NO). Ablation of LGI4 and NO in A23-negative cells significantly attenuates A23-positive cell proliferation and invasion. Our work denotes a driving role of ADAM23-ITH during disease progression, shifting the malignant phenotype from the cellular to the tissue level. Our findings also provide insights for therapeutic intervention, enforcing the need to ascertain ITH to improve cancer diagnosis and therapy.


Subject(s)
ADAM Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Extracellular Matrix Proteins/metabolism , Nitric Oxide/metabolism , ADAM Proteins/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Extracellular Matrix Proteins/genetics , Female , Gene Silencing , Humans , Neoplasm Metastasis , Nerve Tissue Proteins , Tumor Burden , Tumor Microenvironment
14.
Cancer Gene Ther ; 19(8): 558-65, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22699868

ABSTRACT

One of the greatest challenges in urological oncology is renal cell carcinoma (RCC), which is the third leading cause of death in genitourinary cancers. RCCs are highly vascularized and respond positively to antiangiogenic therapy. Endostatin (ES) is a fragment of collagen XVIII that possesses antiangiogenic activity. In this study, we examined the potential of ES-based antiangiogenic therapy to activate tumor-associated endothelial cells in metastatic RCC (mRCC). Balb/c-bearing Renca cells were treated with NIH/3T3-LendSN or, as a control, with NIH/3T3-LXSN cells. The T-cell subsets and lymphocyte populations of tumors, mediastinal lymph nodes and the spleen were assessed by flow cytometry. The expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) was assessed by real-time PCR, flow cytometry and immunohistochemistry analysis. ES gene therapy led to an increase in the percentage of infiltrating CD4-interferon (IFN)-γ cells (P<0.05), CD8-IFN-γ cells (P<0.01) and CD49b-tumor necrosis factor-α cells (P<0.01). In addition, ES therapy caused an increase at the mRNA level of ICAM-1 (1.4-fold; P<0.01) and VCAM-1 (1.5-fold) (control vs treated group; P<0.001). Through flow cytometry, we found a significant increase in the CD34/ICAM-1 cells (8.1-fold; P<0.001) and CD34/VCAM-1 cells (1.6-fold; P<0.05). ES gene therapy induced a significant increase in both T CD4 and CD8 cells in the lymph nodes and the spleen, suggesting that ES therapy may facilitate cell survival or clonal expansion. CD49b cells were also present in increased quantities in all of these organs. In this study, we demonstrate an antitumor inflammatory effect of ES in an mRCC model, and this effect is mediated by an increase in ICAM-1 and VCAM-1 expression in tumor-associated endothelial cells.


Subject(s)
Carcinoma, Renal Cell , Endostatins , Genetic Therapy , Intercellular Adhesion Molecule-1 , Vascular Cell Adhesion Molecule-1 , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Endostatins/genetics , Endostatins/therapeutic use , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
15.
Cell Tissue Res ; 346(3): 427-37, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22120666

ABSTRACT

Galectin-3 (gal-3) is a ß-galactoside binding protein present in multivalent complexes with an extracellular matrix and with cell surface glycoconjugates. In this context, it can deliver a variety of intracellular signals to modulate cell activation, differentiation and survival. In the hematopoietic system, it was demonstrated that gal-3 is expressed in myeloid cells and surrounding stromal cells. Furthermore, exogenous and surface gal-3 drive the proliferation of myeloblasts in a granulocyte-macrophage colony-stimulating factor (GM-CSF)-dependent manner. Here, we investigated whether gal-3 regulates the formation of myeloid bone marrow compartments by studying galectin-3(-/-) mice (gal-3(-/-)) in the C57BL/6 background. The bone marrow histology of gal-3(-/-) mice was significantly modified and the myeloid compartments drastically disturbed, in comparison with wild-type (WT) animals. In the absence of gal-3, we found reduced cell density and diaphyseal disorders containing increased trabecular projections into the marrow cavity. Moreover, myeloid cells presented limited capacity to differentiate into mature myeloid cell populations in gal-3(-/-) mice and the number of hematopoietic multipotent progenitors was increased relative to WT animals. In addition, bone marrow stromal cells of these mice had reduced levels of GM-CSF gene expression. Taken together, our data suggest that gal-3 interferes with hematopoiesis, controlling both precursors and stromal cells and favors terminal differentiation of myeloid progenitors rather than proliferation.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Galectin 3/deficiency , Animals , Cell Differentiation , Galectin 3/genetics , Galectin 3/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Inbreeding , Mice , Mice, Inbred C57BL , Mice, Knockout , Stromal Cells/cytology , Stromal Cells/metabolism
18.
Cell Transplant ; 19(3): 269-77, 2010.
Article in English | MEDLINE | ID: mdl-19951460

ABSTRACT

Endostatin (ES) is a potent inhibitor of angiogenesis and tumor growth. Continuous ES delivery of ES improves the efficacy and potency of the antitumoral therapy. The TheraCyte system is a polytetrafluoroethylene (PTFE) semipermeable membrane macroencapsulation system for implantation of genetically engineered cells specially designed for the in vivo delivery of therapeutic proteins, such as ES, which circumvents the problem of limited half-life and variation in circulating levels. In order to enable neovascularization at the tissues adjacent to the devices prior to ES secretion by the cells inside them, we designed a scheme in which empty TheraCyte devices were preimplanted SC into immunodeficient mice. Only after healing (17 days later) were Chinese hamster ovary cells expressing ES injected into the preimplanted devices. In another model for device implantation, the cells expressing ES where loaded into the immunoisolation devices prior to implantation into the animals, and the TheraCyte were then immediately implanted SC into the mice. Throughout the 2-month study, constant high ES levels of up to 3.7 microg/ml were detected in the plasma of the mice preimplanted with the devices, while lower but also constant levels of ES (up to 2.1 microg/ml plasma) were detected in the mice that had received devices preloaded with the ES-expressing cells. Immunohistochemistry using anti-ES antibody showed reaction within the device and outside it, demonstrating that ES, secreted by the confined recombinant cells, permeated through the membrane and reached the surrounding tissues.


Subject(s)
Cell Transplantation/instrumentation , Cell Transplantation/methods , Cell- and Tissue-Based Therapy/instrumentation , Cell- and Tissue-Based Therapy/methods , Drug Delivery Systems/instrumentation , Endostatins/pharmacokinetics , Animals , CHO Cells , Capsules , Cattle , Cricetinae , Cricetulus , Drug Delivery Systems/methods , Endostatins/blood , Endostatins/therapeutic use , Mice , Mice, SCID
19.
J Leukoc Biol ; 82(2): 300-10, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17456800

ABSTRACT

Galectin-3 (gal-3), a beta-galactoside-binding animal lectin, plays a role in cell-cell and cell-extracellular matrix interactions. Extracellular gal-3 modulates cell migration and adhesion in several physiological and pathological processes. Gal-3 is highly expressed in activated macrophages. Schistosoma mansoni eggs display a large amount of gal-3 ligands on their surface and elicit a well-characterized, macrophage-dependent, granulomatous, inflammatory reaction. Here, we have investigated the acute and chronic phases of S. mansoni infection in wild-type and gal-3(-/-) mice. In the absence of gal-3, chronic-phase granulomas were smaller in diameter, displaying thinner collagen fibers with a loose orientation. Schistosoma-infected gal-3(-/-) mice had remarkable changes in the monocyte/macrophage, eosinophil, and B lymphocyte subpopulations as compared with the infected wild-type mice. We observed a reduction of macrophage number, an increase in eosinophil absolute number, and a decrease in B lymphocyte subpopulation (B220(+/high) cells) in the periphery during the evolution of the disease in gal-3(-/-) mice. B lymphopenia was followed by an increase of plasma cell number in bone marrow, spleen, and mesenteric lymph nodes of the infected gal-3(-/-) mice. The plasma IgG and IgE levels also increased in these mice. Gal-3 plays a role in the organization, collagen distribution, and mobilization of inflammatory cells to chronic-phase granulomas, niches for extramedullary myelopoiesis, besides interfering with monocyte-to-macrophage and B cell-to-plasma cell differentiation.


Subject(s)
Cell Differentiation , Galectin 3/genetics , Lymph Nodes/cytology , Schistosomiasis/immunology , Acute Disease , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Count , Chronic Disease , Crosses, Genetic , Eosinophils/cytology , Eosinophils/physiology , Female , Granuloma/etiology , Granuloma/pathology , Immunoglobulin E/blood , Immunoglobulin G/blood , Immunohistochemistry , Kinetics , Liver/pathology , Lymph Nodes/physiology , Lymphopenia , Macrophages/cytology , Macrophages/physiology , Male , Mesentery/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/physiology , Plasma Cells/cytology , Schistosoma mansoni/immunology , Schistosoma mansoni/pathogenicity , Schistosomiasis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...