Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Microorganisms ; 11(6)2023 May 27.
Article in English | MEDLINE | ID: mdl-37374925

ABSTRACT

Probiotics play a significant role in aquaculture by improving the growth, health, and survival rate of fish against pathogenic organisms. In the present study, we have evaluated the effects of a Lactobacillus rhamnosus (L. rhamnosus) probiotic on growth performance and disease resistance in Oreochromis niloticus (O. niloticus) fingerlings. Four different concentrations of L. rhamnosus (T1: 0.5 × 1010, T2: 1 × 1010, T3: 1.5 × 1010, and T4: 2 × 1010 CFU/kg feed) were administered to fish over a period of three months. L. rhamnosus treated fish revealed a high growth increment as compared to the control, and the values of macromolecules (amino acids, fatty acids, and carbohydrates) varied significantly among the treated and control groups. Levels of thyroid hormones were noted to be high in the probiotic-treated groups. A challenge assay was performed with Aeromonas hydrophila (A. hydrophila). The optimum calculated concentration of probiotics from the growth assay (1.5 × 1010 CFU/kg feed) was used for the challenge assay. Fish were divided into four groups as follows: control (Con), probiotic-treated (PL), infected (I), and infected + probiotic-treated (I + PL) groups. Significant variations in hematological parameters were observed among control and treated groups. Histopathological changes were recorded in infected fish, while the infected + probiotic-treated group showed less deformations indicating the positive effect of the probiotic supplementation. The survival rate of fish was also better in the probiotic-treated group. Based on these findings, we conclude that probiotic supplementation enhances the growth and improves immunity of O. niloticus. Therefore, we propose that probiotics can be used as promising feed supplements for promoting fish production and disease resistance in aquaculture.

2.
Saudi Pharm J ; 31(1): 85-95, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36685298

ABSTRACT

There has been a substantial and consistent rise in the number of clinical trials to develop advanced and potent bispecific antibodies (BsAb) over the past two decades with multiple targets to improve the efficacy or tissue specificity of monoclonal antibodies (mAb) treatment for diseases with multiple determining factors or widely-expressed targets. In this study, we designed and synthesized BsAb AGR2xPD1 targeting extracellular AGR2, a paracrine signal, and PD1, an immune checkpoint protein. Our design is intended to use AGR2 binding to guide PD1 targeting for AGR2+cancer. We used this construction to produce AGR2xPD1 BsAb by generating clonally selected stable 293F cell line with high expression. Applying this BsAb in a T cell-Tumor cell co-culture system showed that targeting both PD1 and AGR2 with this BsAb induces the attachment of TALL-104 (CD8+ T-lymphocytes) cells onto co-cultured H460 AGR2+ Lung tumor cells and significantly reduces migration of H460 cells. T-cell expression of CD8 and IFNγ is also synergistically enhanced by the AGR2xPD1 BsAb treatment in the AGR2+H460 co-culture system. These effects are significantly reduced with AGR2 expression negative WI38 cells. Our results demonstrate that the AGR2xPD1 BsAb could be a potential therapeutic agent to provide better solid tumor targeting and synergetic efficacy for treating AGR2+ cancer by blocking AGR2 paracrine signaling to reduce tumor survival, and redirecting cytotoxic T-cells into AGR2+ cancer cells.

3.
Biochem Biophys Res Commun ; 637: 358-364, 2022 12 31.
Article in English | MEDLINE | ID: mdl-35595575

ABSTRACT

Hemophilia A is a bleeding disorder caused by quantitative or qualitative deficiencies in coagulation factor VIII (FVIII). Low FVIII expression due to its unstable mRNA and binding with immunoglobulin-binding protein (BiP) compromises gene therapy endeavors in hemophilia A. Site-directed mutagenesis have demonstrated an improvement in the expression of FVIII proteins. In this study, a targeted point mutation of Pro at position 290 to Thr (P290T) enhances the in vitro specific activity of B-domain-deleted factor VIII (BDD-FVIII). Hydrodynamic gene delivery of P290T cDNA into FVIII-deficient (FVIII-/-) mice corrected bleeding symptoms. P290T variant resulted in high plasma FVIII coagulant activity 24 h post-gene delivery. Furthermore, bleeding time and average blood loss was significantly reduced in FVIII-/- mice injected with P290T variant, whereas BDD-FVIII and PBS-injected mice experienced prolonged bleeding and excessive blood loss. Histological analysis of the liver biopsies revealed no apparent signs of liver damage. No signs of potential toxicity were seen in mice following mice bodyweights assessment. Altogether, our results demonstrate that the introduction of P290T mutation increases both in vitro and in vivo FVIII coagulant activity, supporting ongoing efforts to develop more effective replacement therapy for hemophilia A.


Subject(s)
Coagulants , Hemophilia A , Animals , Mice , Disease Models, Animal , Factor VIII/genetics , Factor VIII/therapeutic use , Genetic Therapy/methods , Hemophilia A/genetics , Hemophilia A/therapy , Hemorrhage
4.
Biochem Biophys Res Commun ; 573: 86-92, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34399098

ABSTRACT

Anterior gradient 2 (AGR2) is often overexpressed in several types of cancer. AGR2 is cytoplasmic or secreted as an extracellular signal. Intracellular AGR2 properties and role in cancer have been well studied, but its extracellular function is largely unclear. It has been shown that extracellular AGR2 activates endothelial cells and fibroblasts in culture, but the mechanism of AGR2 signaling is not well elucidated. Here, we report that tumor secreted or externally added AGR2 translocates into cytoplasm by endocytosis, binds to ß-catenin and further co-translocates to the nucleus in NIH3T3 fibroblasts. Externally added AGR2 also increased ß-catenin expression, stability, and accumulation in the nucleus in both fibroblasts and cancer cells. External AGR2 rescued the expression of ß-catenin, which was suppressed by EGFR inhibitor AG1478 indicating an alternative pathway to regulate ß-catenin independent of EGFR signal. These effects were abolished when a monoclonal antibody against AGR2 was added to the experiments, confirming the effects are caused by AGR2 only. Putting together, our results show that extracellular AGR2 signaling pathway involves endocytosis mediated cellular translocation, direct binding and regulating ß-catenin nuclear accumulation. It is also a target against tumor initiated AGR2 signaling to form and maintain tumor microenvironment.


Subject(s)
Fibroblasts/metabolism , Mucoproteins/metabolism , Oncogene Proteins/metabolism , beta Catenin/metabolism , Animals , Cell Line , Dimerization , Endocytosis , Humans , Mice
5.
Int Immunopharmacol ; 90: 107155, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33243603

ABSTRACT

IL13Rα2 shows high expression in different types of tumors and can be a target for cancer therapy in humans due to its poor prognosis. The aim of our study is to characterize and investigate the effect of interleukin-13 receptor subunit alpha-2monoclonal antibody mAb15D8 on lung cancer cells in vitro and in vivo by blocking its specific epitope in IL13Rα2 antigen. The mAb15D8 blocking epitope was analyzed through the mutagenesis of IL13Rα2 and confirmed with western blot. We found that the IL13Rα2 epitope recognized by mAb15D8 antibody is a new binding site localized in the fibronectin-III domain-1 of IL13Rα2 antigen. Moreover, the mAb15D8 obviously reduced cell proliferation, migration of H460, A549, SKOV3, and B16F10 cells. Treatment with mAb15D8 significantly reduced the H460 xenograft tumor formation and growth in nude mice and inhibited B16F10 tumor metastasis and increased survival in C57BL/6 mice. Pharmacokinetic and toxicological analysis demonstrated the safety of mAb15D8 as a potential therapeutic agent. We developed a novel mouse monoclonal antibody against IL13Rα2 which binds to specific epitope on IL13Rα2 antigen. In vivo treatment with the antibody significantly reduced tumor growth and lung metastasis and prolonged survival. These results suggest mAb15D8 antibody as a potential therapeutic agent for cancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Interleukin-13 Receptor alpha2 Subunit/genetics , Interleukin-13 Receptor alpha2 Subunit/immunology , Interleukin-13 Receptor alpha2 Subunit/metabolism , Lung Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/toxicity , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epitope Mapping , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Xenograft Model Antitumor Assays
6.
Curr Pharm Des ; 2020 09 09.
Article in English | MEDLINE | ID: mdl-32912123

ABSTRACT

The article has been withdrawn by the editorial office of the journal Current Pharmaceutical Design, due to major linguistic inconsistencies.Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused.The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorial-policies-main.php BENTHAM SCIENCE DISCLAIMER: It is a condition of publication that manuscripts submitted to this journal have not been published and will not be simultaneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been published elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and by submitting the article for publication the authors agree that the publishers have the legal right to take appropriate action against the authors, if plagiarism or fabricated information is discovered. By submitting a manuscript, the authors agree that the copyright of their article is transferred to the publishers if and when the article is accepted for publication.

7.
Cell Adh Migr ; 13(1): 332-344, 2019 12.
Article in English | MEDLINE | ID: mdl-31710263

ABSTRACT

The most prominent cancer-associated fibroblasts (CAFs) in tumor stroma is known to form a protective structure to support tumor growth. Anterior gradient-2 (AGR2), a tumor secretory protein is believed to play a pivotal role during tumor microenvironment (TME) development. Here, we report that extracellular AGR2 enhances fibroblasts elongation and migration significantly. The early stimulation of RhoA showed the association of AGR2 by upregulation of G1-S phase-regulatory protein cyclin D1 and FAK phosphorylation through fibroblasts growth factor receptor (FGFR) and vascular endothelial growth factor receptor (VEGFR). Our finding indicates that secretory AGR2 alters fibroblasts elongation, migration, and organization suggesting the secretory AGR2 as a potential molecular target that might be responsible to alter fibroblasts infiltration to support tumor growth.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Mucoproteins/metabolism , Neoplasms/pathology , Oncogene Proteins/metabolism , Tumor Microenvironment/physiology , rhoA GTP-Binding Protein/metabolism , 3T3 Cells , Animals , Cell Cycle/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Culture Media, Conditioned/pharmacology , Cyclin D1/biosynthesis , Focal Adhesion Kinase 1/metabolism , Humans , MCF-7 Cells , Mice , Phosphorylation , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism
8.
Cancer Lett ; 449: 125-134, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30685412

ABSTRACT

Increased drug resistance and acute side effects on normal organs are the major disadvantages of traditional cancer chemotherapy and radiotherapy. This has increased the focus on targeted therapeutic strategies such as monoclonal antibody-based cancer therapies. The major advantage of antibody-based therapies is the specific inhibition of cancer-related targets, with reduced off-target side effects. Anterior gradient-2 (AGR2) is a prometastatic and proangiogenic tumor marker that is overexpressed in multiple cancers. Therefore, anti-AGR2 antibodies may be potential therapeutic agents for treating different cancers. In the present study, we examined a novel anti-AGR2 monoclonal antibody mAb18A4 and found that this antibody inhibited lung cancer progression and metastasis without exerting any adverse side effects on the major organs and blood in mice. Moreover, we found that mAb18A4 activated p53 pathway and attenuated ERK1/2-MAPK pathway. Furthermore, mAb18A4-treated cancer cell lines showed attenuated proliferation and colony formation, enhanced apoptosis, increased p53 expression, and reduced phosphorylated ERK1/2 expression. Treatment with mAb18A4 significantly reduced tumor size and suppressed tumor metastasis in and increased the survival of different xenograft tumor models. In addition, mAb18A4 potently suppressed AGR2-induced angiogenesis. Results of pharmacokinetic and toxicological analyses confirmed the safety of mAb18A4 as an antitumor treatment.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mucoproteins/antagonists & inhibitors , Oncogene Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , A549 Cells , Animals , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/metabolism , Mice , Signal Transduction/drug effects , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL