Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 245
Filter
1.
Brasília; CONITEC; mar. 2022.
Non-conventional in Portuguese | LILACS, BRISA/RedTESA | ID: biblio-1368853

ABSTRACT

INTRODUÇÃO: O baricitinbe, um imunomodulador que atua sobre a atividade da IL-6 (citocina pró-inflamatória), pode representar uma estratégia para o tratamento de pacientes com COVID-19 que tiveram comprometimento pulmonar devido a resposta hiperinflamátoria desencadeada pela tempestade de citocinas característica na infecção causada pelo vírus SARS-COV2. TECNOLOGIA: Baricitinibe (Olumiant®). EVIDÊNCIAS CLÍNICAS: Para seleção das evidências clínicas foi conduzida uma revisão sistemática da literatura em busca de ensaios clínicos randomizados (ECR), estudos observacionais (mundo real) e revisões sistemáticas que avaliassem os efeitos do baricitinibe como monoterapia ou associado aos cuidados usuais - definidos aqui como 'terapia padrão' (corticoesteróides sistêmicos, anticoagulantes, antimicrobianos/antivirais) no tratamento de pacientes adultos com COVID-19, hospitalizados e que necessitam de suplementação de oxigênio (máscara ou cateter nasal, alto fluxo de oxigênio ou ventilação não invasiva). As buscas eletrônicas foram realizadas nas bases de dados: the Cochrane Library, MEDLINE via Pubmed, Literatura Latino-Americana e do Caribe em Ciências da Saúde (LILACS), EMBASE e Centre for Reviews and Dissemination (CRD). O risco de viés dos estudos primários incluídos foi avaliado pelas ferramentas Risk of Bias versão 2 da Cochrane (para ECR) ou ROBINS-I (para estudos observacionais), e a qualidade metodológica das revisões sistemáticas foi avaliada pela ferramenta AMSTAR-2. A qualidade da evidência foi avaliada pelo sistema GRADE. Seis artigos foram incluídos na presente revisão, sendo dois deles referentes a um ensaio clínico randomizado (ECR), um estudo observacional e três revisões sistemáticas com meta-análise (RSMA), sendo uma


Subject(s)
Humans , Oxygen Inhalation Therapy/instrumentation , Janus Kinases/antagonists & inhibitors , Noninvasive Ventilation/instrumentation , SARS-CoV-2/drug effects , COVID-19/drug therapy , Immunosuppressive Agents/antagonists & inhibitors , Unified Health System , Brazil , Cost-Benefit Analysis/economics , Inpatients
2.
Carbohydr Polym ; 251: 116930, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33142551

ABSTRACT

The aim of this study was to investigate the surface morphological features and in vivo immunomodulatory activities of a hetero polysaccharide fraction (HEP-W) from Hericium erinaceus. SEM and AFM images revealed that HEP-W displayed a flexible random coil conformation, and these flexible winding chains further formed continuous fiber network structure. Meanwhile, Congo red assay and XRD further proved that HEP-W mainly exhibited amorphous structure with non-triple-helical conformation in solution. In vivo immunomodulatory experiments demonstrated that HEP-W possessed protective effects against cyclophosphamide-induced immunosuppression in mice by significantly enhancing immune organ index, splenocyte proliferation, NK cell activity, IL-2 production as well as improving the macrophage phagocytosis. These findings suggest that HEP-W could be explored as a natural and effective immunomodulatory agent.


Subject(s)
Cyclophosphamide/adverse effects , Cyclophosphamide/antagonists & inhibitors , Fungal Polysaccharides/chemistry , Fungal Polysaccharides/pharmacology , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/antagonists & inhibitors , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Carbohydrate Conformation , Congo Red , Female , Fungal Polysaccharides/ultrastructure , Hericium/chemistry , Interleukin-2/blood , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Leukocytes/drug effects , Leukocytes/immunology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred BALB C , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Phagocytosis/drug effects , Surface Properties , X-Ray Diffraction
3.
Prostate ; 79(14): 1611-1621, 2019 10.
Article in English | MEDLINE | ID: mdl-31348843

ABSTRACT

BACKGROUND: The transcription factor signal transducer and activator of transcription 3 (STAT3) is implicated in cancer drug resistance, metastasis, and immunosuppression and has been identified as a promising therapeutic target for new anticancer drugs. Myeloid-derived suppressor cells (MDSCs) play a major role in the suppression of antitumor immunity and STAT3 is involved in the accumulation, generation, and function of MDSCs. Thus, targeting STAT3 holds the potential of reversing immunosuppression in cancer. This study aims to investigate the effect of the small molecule STAT3 inhibitor galiellalactone on prostate cancer cell- induced generation of MDSCs from monocytes and the effect on immunosuppressive factors and inflammatory cytokines. METHODS: Primary human monocytes were cocultured with prostate cancer cells (DU145, PC3, and LNCaP-IL6) or with conditioned medium (CM) from prostate cancer cells in the presence or absence of the STAT3 inhibitor galiellalactone. Monocytes were analyzed by flow cytometry for an MDSC-like phenotype (CD14+ HLA-DR-/lo ). The secretion and gene expression of immunosuppressive factors and inflammatory cytokines from prostate cancer cells and monocytes were investigated. RESULTS: Galiellalactone blocked the prostate cancer cell-induced generation of MDSC-like monocytes with an immunosuppressive phenotype ex vivo. Monocytes cultured with CM from prostate cancer cells showed increased expression of phosphorylated STAT3. Prostate cancer cells increased the expression of interleukin1ß (IL1ß), IL10, and IL6 in monocytes which was inhibited by galiellalactone. In addition, galiellalactone decreased indoleamine 2,3-dioxygenase gene expression in monocytes. Galiellalactone reduced the levels of IL8 and granulocyte macrophage-colony stimulating factor in prostate cancer cells per se. CONCLUSION: The STAT3 inhibitor galiellalactone may prevent the prostate cancer cell-induced generation of MDSCs and reverse the immunosuppressive mechanisms caused by the interplay between prostate cancer cells and MDSCs. This is a potential new immunotherapeutic approach for the treatment of prostate cancer.


Subject(s)
Carcinogens/antagonists & inhibitors , Immunosuppressive Agents/antagonists & inhibitors , Lactones/pharmacology , Myeloid-Derived Suppressor Cells/drug effects , Prostatic Neoplasms/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Carcinogenesis/drug effects , Carcinogens/metabolism , Coculture Techniques , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Humans , Immunosuppressive Agents/metabolism , Male , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured
4.
Am J Chin Med ; 46(8): 1879-1897, 2018.
Article in English | MEDLINE | ID: mdl-30518233

ABSTRACT

A variety of products have been developed with red ginseng (RG, the steamed roots of Panax ginseng Meyer). To clarify the immunomodulating effects of water-extracted RG (wRG), 50% ethanol-extracted RG (eRG), enzyme-treated eRG (ERG) and probiotic-fermented eRG (FRG), we examined their immunopotentiating and immunosuppressive effects in mice with cyclophosphamide (CP)-induced immunosuppression (CI) or 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis (TC). Oral administration of RG in CI mice significantly increased blood IFN- γ levels. Treatment with RG also increased the tumoricidal effects of CI mouse splenic cytotoxic T (Tc) and NK cells against YAC-1 cells. Treatment with RGs, in particular FRG and wRG, significantly increased Th1 cell differentiation. Treatment with RG except wRG increased Treg cell differentiation. However, wRG alone increased IL-6 and IL-17 expression in the colon of CI mice. Furthermore, RG alleviated colitis in TC mice. FRG most potently suppressed TNBS-induced colon shortening, NF- κ B activation and TNF- α and IL-17 expression and increased IL-10 expression. RGs inhibited TNF- α expression and increased IL-10 expression in lipopolysaccharide-stimulated primary macrophages in vitro while the differentiation of splenic T cells into type 1 T (Th1) and regulatory T (Treg) cells was increased by FRG in vitro. In conclusion, FRG can alleviate immunosuppression and inflammation by inhibiting macrophage activation and regulating Th1 and Treg cell differentiation.


Subject(s)
Adjuvants, Immunologic , Cell Differentiation/drug effects , Colitis/drug therapy , Cyclophosphamide/antagonists & inhibitors , Fermentation , Immunosuppressive Agents/antagonists & inhibitors , Macrophage Activation/drug effects , Panax/chemistry , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , T-Lymphocytes/physiology , Trinitrobenzenesulfonic Acid/adverse effects , Administration, Oral , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Male , Mice, Inbred BALB C , Plant Extracts/administration & dosage , Plant Extracts/isolation & purification
5.
Drug Metab Pharmacokinet ; 33(5): 232-239, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30241694

ABSTRACT

The cellular uptake of mizoribine (MZR), an immunosuppressant, and metabolism of MZR to MZR-5'- monophosphate (MZRP), an active metabolite, were evaluated in L5178Y-R mouse lymphoma cells and peripheral blood mononuclear cells (PBMCs) of rats and kidney transplant recipients (KTRs, n = 22). Real-time PCR analysis revealed the expression of ENT1 and ENT2 mRNAs, but not of CNTs, in L5178Y-R cells and rat's PBMCs. In L5178Y-R cells, the uptake of MZR was suppressed by adenosine, a substrate for ENT1 and ENT2, but not by 5-(4-nitrobenzyl)-6-thioinosine (0.1 µM), an ENT1 inhibitor. Saturable metabolism of MZR to MZRP was observed. In rats, peak plasma concentrations of MZR and peak concentrations of MZR and MZRP in PBMCs were observed 3 h after oral administration. MZR disappeared from PBMCs in parallel with plasma MZR, but the disappearance of MZRP from PBMCs appeared to be slow. In KTRs, the mean plasma concentration of MZR 3-4 h after ingestion was 3.14 µg/ml and the mean MZRP concentration in PBMCs was 16.8% of MZR, reflecting the involvement of ENT in the uptake of MZR. A linear relationship was observed between plasma MZR concentrations ranging from 1 to 6 µg/ml and PBMC's MZRP concentrations ranging from 90 to 200 ng/ml.


Subject(s)
Immunosuppressive Agents/metabolism , Kidney Transplantation , Leukemia L5178/pathology , Leukemia L5178/therapy , Leukocytes, Mononuclear/metabolism , Ribonucleosides/metabolism , Adenosine/pharmacology , Administration, Oral , Animals , Immunosuppressive Agents/antagonists & inhibitors , Leukemia L5178/metabolism , Leukocytes, Mononuclear/drug effects , Male , Mice , Mice, Inbred ICR , Rats , Rats, Sprague-Dawley , Ribonucleosides/antagonists & inhibitors
6.
Lab Invest ; 97(11): 1271-1281, 2017 11.
Article in English | MEDLINE | ID: mdl-28759009

ABSTRACT

We previously reported that long-term treatment with a calcineurin inhibitor impairs autophagy process in pancreatic beta cells. This study investigated the effect of Korean red ginseng extract (KRGE) on autophagy modulated by oxidative stress. In mice with tacrolimus (Tac)-induced diabetes mellitus, KRGE alleviated islet dysfunction and decreased oxidative stress and autophagic vacuoles. In vitro, KRGE decreased autophagosome formation and attenuated lysosomal degradation, accompanied by improved beta cell viability and insulin secretion. Addition of 3-methyladenine (3-MA), an inhibitor of autophagosomes, to KRGE further improved cell viability and insulin secretion, and bafilomycin A (BA), an inhibitor of lysosomal function, reduced the effects of KRGE. At the subcellular level, Tac caused mitochondrial dysfunction (impaired mitochondrial oxygen consumption, ATP production, and increased reactive oxygen species production). But KRGE improved these parameters. The effect of KRGE on mitochondrial function enhanced by 3-MA but decreased by BA, suggesting a causal relationship between KRGE effect and autophagy modulation in Tac-induced mitochondrial dysfunction. These findings indicate that KRGE modulates autophagy favorably by reducing Tac-induced oxidative stress, and this effect is closely associated with improvement of mitochondrial function.


Subject(s)
Antioxidants/therapeutic use , Autophagy , Diabetes Mellitus/prevention & control , Dietary Supplements , Insulin-Secreting Cells/metabolism , Panax/chemistry , Plant Extracts/therapeutic use , Animals , Antioxidants/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/pathology , Autophagosomes/ultrastructure , Autophagy/drug effects , Calcineurin Inhibitors/adverse effects , Calcineurin Inhibitors/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/antagonists & inhibitors , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/ultrastructure , Male , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/ultrastructure , Oxidative Stress/drug effects , Plant Extracts/metabolism , Plant Roots/chemistry , Random Allocation , Rats , Tacrolimus/adverse effects , Tacrolimus/antagonists & inhibitors
7.
Int J Biol Macromol ; 95: 1216-1222, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27825995

ABSTRACT

The present study evaluated the ameliorative effects of Trametes orientalis polysaccharide (TOP-2) against cyclophosphamide (CP) induced toxicity in mice. Intraperitoneal administration of TOP-2 not only effectively increased the thymus, spleen, heart, liver, and kidney indices, but also significantly enhanced the phagocytic activities of macrophages and splenocyte proliferation, dose-dependently. The lowered nitric oxide level of macrophages after CP treatment was elevated by TOP-2 administration. Impaired splenic natural killer cells and cytotoxic T lymphocytes activities were remarkably enhanced by TOP-2. Furthermore, the levels of interleukin-2, interferon-γ, Immunoglobulin A, immunoglobulin G, and immunoglobulin M were notably reduced by CP, while TOP-2 abolished these effects. TOP-2 could also effectively increase the total antioxidant capacity, superoxidase dismutase, catalase and glutathione peroxidase activities, and inhibit the increase in malondialdehyde level. These results indicate that TOP-2 may be of therapeutic value in ameliorating the immunosuppression and oxidative stress caused by CP treatment, through its immunomodulatory and antioxidant potential.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cyclophosphamide/antagonists & inhibitors , Fungal Polysaccharides/pharmacology , Immunosuppressive Agents/antagonists & inhibitors , Trametes/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Antioxidants/chemistry , Antioxidants/isolation & purification , Catalase/immunology , Catalase/metabolism , Cyclophosphamide/adverse effects , Female , Fungal Polysaccharides/chemistry , Fungal Polysaccharides/isolation & purification , Glutathione Peroxidase/immunology , Glutathione Peroxidase/metabolism , Heart/drug effects , Immunosuppression Therapy , Immunosuppressive Agents/adverse effects , Kidney/drug effects , Kidney/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Liver/drug effects , Liver/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Malondialdehyde/immunology , Malondialdehyde/metabolism , Mice , Oxidative Stress/drug effects , Phagocytosis/drug effects , Spleen/drug effects , Spleen/immunology , Superoxide Dismutase/immunology , Superoxide Dismutase/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Thymus Gland/drug effects , Thymus Gland/immunology
8.
Adv Chronic Kidney Dis ; 23(5): 301-305, 2016 09.
Article in English | MEDLINE | ID: mdl-27742384

ABSTRACT

Although calcineurin inhibitor drugs have been the mostly used therapy in modern immunosuppression in kidney transplantation, their effect on kidney allograft dysfunction has been suboptimal as far as preservation of kidney function is concerned. Additionally, there are metabolic and other nonmetabolic effects including increased risk of malignancy that has necessitated the use of mammalian target of rapamycin inhibitors to reduce exposure to calcineurin inhibitors. Mammalian target of rapamycin inhibitors, both sirolimus and everolimus, have been studied in several trials to facilitate preservation of kidney function with variable effects on kidney allograft function and immunogenicity. Preservation of kidney function is increasingly becoming the mainstay of immunosuppression not only in kidney transplantation, but also in extrakidney transplantation. The best kidney outcomes have been reported in calcineurin inhibitor withdrawal studies using mammalian target of rapamycin inhibitors, in kidney transplant recipients with stable kidney function. This review article summarizes data from several studies in which mammalian target of rapamycin inhibitors have been used to reduce exposure to or withdraw calcineurin inhibitors in an attempt to preserve kidney function.


Subject(s)
Acute Kidney Injury/prevention & control , Calcineurin Inhibitors/therapeutic use , Graft Rejection/prevention & control , Immunosuppression Therapy/methods , Immunosuppressive Agents/antagonists & inhibitors , Kidney Transplantation , Sirolimus/antagonists & inhibitors , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Animals , Graft Rejection/complications , Graft Rejection/immunology , Humans
9.
Biochem Pharmacol ; 117: 1-9, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27038657

ABSTRACT

A previously studied immunosuppressive cytokine, Soluble Immune Response Suppressor (SIRS), may have relevance to current studies of immune suppression in a variety of human disease states. Despite extensive efforts using experimental models, mainly in mice, much remains to be discovered as to how autoimmune cells in mice and humans escape normal regulation and, conversely, how tumor cells evade evoking an immune response. It is the contention of this commentary that the literature pre-2000 contain results that might inform current studies. The broadly immunosuppressive protein, SIRS, was studied extensively from the 1970s to 1990s and culminated in the determination of the n-terminal 21mer sequence of this 15kDa protein which had high homology to the short neurotoxins from sea snakes, that are canonical members of the three finger neurotoxin superfamily (3FTx). It was not until 2007 that the prophylactic administration of the synthetic N-terminal peptide of the SIRS 21mer, identical to the published sequence, was reported to inhibit or delay the development of two autoimmune diseases in mice: experimental allergic encephalomyelitis (EAE) and type I diabetes (T1D). These findings were consistent with other studies of the 3FTx superfamily as important probes in the study of mammalian pharmacology. It is the perspective of this commentary that SIRS, SIRS peptide and the anti-peptide mAb, represent useful, pharmacologically-active probes for the study of the immune response as well as in the potential treatment of autoimmune, inflammatory diseases and cancer.


Subject(s)
Autoimmune Diseases/drug therapy , Immunosuppressive Agents/therapeutic use , Models, Molecular , Suppressor Factors, Immunologic/therapeutic use , Algorithms , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Computational Biology , Expert Systems , Humans , Immunomodulation/drug effects , Immunosuppression Therapy/methods , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Neurotoxins/chemistry , Neurotoxins/toxicity , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Protein Conformation , Sequence Homology, Amino Acid , Snake Venoms/chemistry , Snake Venoms/toxicity , Suppressor Factors, Immunologic/antagonists & inhibitors , Suppressor Factors, Immunologic/chemistry , Suppressor Factors, Immunologic/pharmacology
10.
Life Sci ; 152: 1-13, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26997472

ABSTRACT

Aflatoxin B1 (AFB1) is immunotoxic to animals and is a suspected immunosuppressant in humans. ß-1,3-Glucan (BG) consists of glucose polymers and has a variety of stimulatory effects on the immune system. In this study, we investigated the role of BG on the expression of phenotypic markers and cytokine secretion in mice exposed to AFB1. We treated animals with BG (150mg/kg, p.o., once daily) for 7days beginning at the onset of AFB1 exposure. Exposure of animals to AFB1 alone (1250µg/kg, p.o, once daily) for 7days resulted in a decrease in the percentages of lymphocyte subsets (CD4(+), GITR(+), CD8(+), TCR ß(+), CD3(+), Foxp3(+), CD4(+)Foxp3(+), and CD127(+)) as compared to an normal control (NC). However, both BG alone and BG given in conjunction with exposure to AFB1 significantly increased the percentages of these lymphocyte subsets in blood. We also observed that mice exposed to AFB1 showed reduced IL-2, TNF-α, IL-17, and IFN-γ production in the spleen and serum. In contrast, oral administration of BG alone and in conjunction with AFB1 exposure augmented the levels of these cytokines. Moreover, this finding was confirmed through RT-PCR and western blot analysis of mRNA and protein expression in the spleen. Altogether, it can be concluded from these studies that BG enhances the responses of lymphocyte subsets, including cytokine production, even when given following exposure to AFB1 immunotoxin. These data demonstrate that BG carries out its immunomodulating activity by regulating cytokine production. Our findings also provide a direction for development of specific immunomodulating therapy.


Subject(s)
Aflatoxin B1/antagonists & inhibitors , Immunity/drug effects , Immunologic Factors/pharmacology , Immunosuppressive Agents/antagonists & inhibitors , beta-Glucans/pharmacology , Animals , Cytokines/blood , Cytokines/metabolism , Forkhead Transcription Factors/biosynthesis , Immunity, Cellular/drug effects , Lymphocyte Count , Lymphocyte Subsets/drug effects , Male , Mice , Mice, Inbred BALB C , Protein Synthesis Inhibitors/pharmacology , Spleen/drug effects , Spleen/metabolism , Transcription Factor RelA/biosynthesis
11.
Carbohydr Polym ; 142: 259-67, 2016 May 20.
Article in English | MEDLINE | ID: mdl-26917398

ABSTRACT

An exopolysaccharide (EPS) was fractionated from fermentation media of a Cordyceps sinensis fungus (Cs-HK1) by ethanol precipitation at 2/5 volume ratio of ethanol/media. Its structural characteristics were elucidated by FT-IR, GC, GC-MS, 1D and 2D NMR combined with periodate oxidation, Smith degradation, partial acid hydrolysis, and methylation analysis. Furthermore, the immunomodulatory activity of EPS was evaluated by the model of cyclophosphamide-induced immunosuppression. The results from monosaccharide composition and partial acid hydrolysis indicated that EPS almost consisted of glucose excluding a trace amount of mannose. GC-MS and NMR analysis further confirmed EPS had a linear backbone of (1→3)-ß-D-glucopyranosyl residues with a single (1→6)-ß-D-glucopyranosyl side-branching unit for every three ß-D-glucopyranosyl residues, showing a comb-like ß-D-glucan with short and intensive branches, which was responsible for high viscosity. Moreover, EPS could significantly enhance immune organs and stimulate the release of major cytokines TNF-α and INF-γ, suggesting that EPS exhibited protective effect in immunocompromised mice.


Subject(s)
Cordyceps/chemistry , Cyclophosphamide/antagonists & inhibitors , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Immunosuppressive Agents/antagonists & inhibitors , beta-Glucans/chemistry , beta-Glucans/pharmacology , Animals , Cyclophosphamide/pharmacology , Female , Hydrolysis , Immunologic Factors/isolation & purification , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Interferon-gamma/blood , Interleukin-10/blood , Mice , Spleen/drug effects , Spleen/immunology , Thymus Gland/drug effects , Thymus Gland/immunology , Tumor Necrosis Factor-alpha/blood , beta-Glucans/isolation & purification
12.
J Physiol Biochem ; 72(2): 133-44, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26746208

ABSTRACT

Tacrolimus (TAC), a calcineurin inhibitor (CNI), is clinically used as an immunosuppressive agent in the transplant recipient; however, the use of TAC is greatly limited by its nephrotoxicity and hepatotoxicity. Mycophenolate mofetil (MMF), an inhibitor of the purine synthesis, has been used in combination with many immunosuppressive drugs such as TAC. The association TAC/MMF was used in organ transplantation to increase the efficiency and reduce acute rejection rates, but the effects of MMF on TAC-induced kidney and liver injuries are still not well investigated. The aims of this study are to explore whether MMF co-administration with TAC has a renoprotective and hepatoprotective effect against TAC-induced renal and hepatic injuries and to check the implication of oxidative stress in the MMF's possible protective effect. Our results showed that MMF (at 50 mg kg(-1) body weight (b.w.)) restored creatinine, in addition to increased AST and ALT levels by TAC (at 60 mg kg(-1) b.w.). Furthermore, MMF decreased DNA damage induced by TAC in the kidney and liver of rats as assessed by comet assay. This renoprotective and hepatoprotective effect of MMF was associated with an antioxidant effect. In fact, MMF co-treatment with TAC decreased oxidative damage induced by TAC. It reduced malondialdehyde (MDA) and protein carbonyl (PC) levels as well as catalase and superoxide dismutase (SOD) activities. We conclude that the co-administration MMF with TAC protect liver and kidney against TAC toxicity via an antioxidant process.


Subject(s)
Calcineurin Inhibitors/adverse effects , Chemical and Drug Induced Liver Injury/prevention & control , Immunosuppressive Agents/adverse effects , Mycophenolic Acid/therapeutic use , Protective Agents/therapeutic use , Renal Insufficiency/prevention & control , Tacrolimus/adverse effects , Animals , Biomarkers/blood , Biomarkers/metabolism , Calcineurin Inhibitors/administration & dosage , Calcineurin Inhibitors/chemistry , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , DNA Damage/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/metabolism , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/antagonists & inhibitors , Kidney/drug effects , Kidney/metabolism , Kidney/physiopathology , Liver/drug effects , Liver/metabolism , Liver/physiopathology , Male , Oxidative Stress/drug effects , Protective Agents/administration & dosage , Protein Carbonylation/drug effects , Random Allocation , Rats, Wistar , Renal Insufficiency/chemically induced , Renal Insufficiency/metabolism , Renal Insufficiency/physiopathology , Tacrolimus/administration & dosage , Tacrolimus/antagonists & inhibitors
13.
Lab Invest ; 95(10): 1174-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26237274

ABSTRACT

Accumulating evidence shows that a gut-released hormone, the glucagon-like peptide-1 (GLP-1), has not only a glucose-lowering effect but also a renoprotective effect against kidney injury. In this study, we investigated whether a dipeptidyl peptidase (DPP) IV inhibitor has a protective effect against tacrolimus-induced renal injury. Rats were treated with tacrolimus (1.5 mg/kg, subcutaneously) and the DPP IV inhibitor MK0626 (10 or 20 mg/kg, oral gavage) for 4 weeks. MK0626 treatment attenuated tacrolimus-induced renal dysfunction, tubulointerstitial fibrosis, and arteriolopathy. Moreover, these improvements were accompanied by a reduction in oxidative stress and apoptosis. MK0626 treatment increased the blood level of GLP-1 and the level of its receptor in tissue sections but did not alter the levels of other DPP IV substrates, such as neuropeptide Y and the stromal cell-derived factor-1. These data suggest that DPP IV inhibition has an important role in the renoprotection against tacrolimus-induced nephrotoxicity via antioxidative and antiapoptotic effects and preservation of the GLP-1 system.


Subject(s)
Calcineurin Inhibitors/adverse effects , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Immunosuppressive Agents/adverse effects , Kidney/drug effects , Renal Insufficiency, Chronic/prevention & control , Tacrolimus/adverse effects , Triazoles/therapeutic use , Animals , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Apoptosis/drug effects , Biomarkers/blood , Biomarkers/metabolism , Calcineurin Inhibitors/chemistry , Diet, Sodium-Restricted/adverse effects , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Fibrosis , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/antagonists & inhibitors , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/metabolism , Immunosuppressive Agents/antagonists & inhibitors , Kidney/blood supply , Kidney/metabolism , Kidney/pathology , Male , Oxidative Stress/drug effects , Random Allocation , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Tacrolimus/antagonists & inhibitors , Triazoles/administration & dosage
14.
Environ Toxicol Pharmacol ; 39(3): 1262-75, 2015 May.
Article in English | MEDLINE | ID: mdl-26026872

ABSTRACT

Cyclophosphamide (CP) is a cytotoxic drug that can suppress both humoral and cellular immunity. Combining traditional medicinal herbs and chemotherapy drugs are used to improve immunity and quality of life performance status. In this paper, the effects of plant extracts, active components and their derivatives on immunosuppression of CP are discussed. Appropriate keywords were used to search through PubMed, Google Scholar, and Sciverse. All relevant results published from 1990 to date were chosen for final review. Over 50 references were found in which plant extracts, active components and their derivatives have been tested for their immune protective effects against CP-induced immune toxicity. Although there are several plants shown to be effective in animal models, no study was carried out on human subjects. According to the results; we can claim that plants and their active ingredients are good candidates for alternative adjuvant chemotherapy in reducing the immunotoxicity of CP.


Subject(s)
Cyclophosphamide/antagonists & inhibitors , Immunosuppressive Agents/antagonists & inhibitors , Plant Extracts/pharmacology , Animals , Databases, Bibliographic , Humans , Phytotherapy
15.
Appl Biochem Biotechnol ; 176(3): 742-57, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25900435

ABSTRACT

Cancer is the leading cause of death worldwide. Cyclophosphamide (CTX) is commonly used as anticancer drug which causes toxicity by its reactive metabolites such as acroline and phosphoramide mustard. In this study, Cuscuta chinensis (C. chinensis) (family: Convolvulaceae) was assessed for ability to restore mice against CTX-induced toxicity. Coadministration of C. chinensis extract (10 mg/kg BW, IP, daily) for ten consecutive days reduced CTX-induced (25 mg/kg BW, IP, daily) toxicity. Treatment with C. chinensis extract significantly (p < 0.01) increased the relative organ weight and body weight. Moreover, administration of C. chinensis extract significantly increased bone marrow cellulatity and α-esterase activity in CTX-treated mice which suggested its protective role on the hematopoietic system. The GSH content was drastically reduced by CTX administration in urinary bladder which was enhanced by treatment with C. chinensis extract, indicating that preventing acroline-mediated tissue damage or cell toxicity and also the extract decreased the urinary bladder nitric oxide (NO) level which proves recovery over urinary tract injury associated with CTX treatment. The administration of C. chinensis extract decreased serum urea, creatinine, and bilirubin levels when compared to CTX-alone-treated group. Histopathological analysis of the urinary bladder of CTX-alone-treated group showed necrotic damage whereas the C. chinensis-treated group showed normal bladder architecture. The above data clearly demonstrates chemoprotective role of C. chinensis against CTX-induced toxicities by regulating antioxidant and inflammatory mediators.


Subject(s)
Cuscuta/chemistry , Cyclophosphamide/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Plant Extracts/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Urine/chemistry , Animals , Antineoplastic Agents/adverse effects , Bilirubin/blood , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Creatinine/blood , Cyclophosphamide/antagonists & inhibitors , Esterases/metabolism , Glutathione/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/antagonists & inhibitors , Male , Methanol/chemistry , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Organ Size/drug effects , Plant Extracts/chemistry , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/blood , Urea/blood , Urinary Bladder/drug effects
16.
Eksp Klin Farmakol ; 78(12): 15-7, 2015.
Article in Russian | MEDLINE | ID: mdl-27051922

ABSTRACT

We have studied the effect of phenotropil on the level of cytokines IL-1, IL-4, and IL-6 under conditions of immunosuppression induced by cyclophosphamide (150 mg/kg, single i.p. injection) and under immune system hyperreactivity caused by lipopolysaccharide (LPS of Pseudomonas aeruginosa, 100 gg/kg, single i.p. injection). The experiments were performed on 48 Wistar male rats aged 6-8 months. Each series was carried out according to similar schedule onree groups of animals: group 1, intact animals; group 2 received cyclophosphamide (series 1) or LPS (series 2); group 3 received phenotropil on the background of immunomodulators. Phenotropil (synthesized at the Department of Organic Chemistry of the A. I. Herzen St. Petersburg Pedagogical University) was administered in a dose of 25 mg/kg (i.p.) for 5 days, starting 2 days before the introduction of immune system activity modulatos. The levels of serum cytokines IL-1ß, IL-4, and IL-6 were determined by ELISA "sandwich" with the use of monoclonal antibodies (ELISA kits of BenderMedsystems company; instrument Uniplan AIRF-01 of Picon company, Moscow) at a wavelength of 450 nm. In case of immunodeficiency caused by cyclophosphamide, there was a significant decrease in IL-1ß, IL-4 and IL-6, but the introduction of phenotropil largely restored their levels. Upon stimulation with LPS, the immune response showed inc- rease in pro-inflammatory cytokines IL-ß, IL-6 and decrease in IL-4. Under these conditions, phenotropil restored the normal levels of IL-1ß, IL-6 and caused an increase in the level of IL-4.


Subject(s)
Immunologic Factors/pharmacology , Interleukin-1beta/blood , Interleukin-4/blood , Interleukin-6/blood , Piracetam/analogs & derivatives , Animals , Cyclophosphamide/antagonists & inhibitors , Cyclophosphamide/pharmacology , Drug Administration Schedule , Immunity, Innate/drug effects , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/pharmacology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Injections, Intraperitoneal , Interleukin-1beta/biosynthesis , Interleukin-4/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides , Male , Piracetam/pharmacology , Pseudomonas aeruginosa/chemistry , Rats , Rats, Wistar , Th1-Th2 Balance/drug effects
17.
Int J Pharm ; 474(1-2): 1-5, 2014 Oct 20.
Article in English | MEDLINE | ID: mdl-25091376

ABSTRACT

Edelfosine, an alkyl-lysophospholipid antitumor drug with severe side-effects, has previously been encapsulated into lipid nanoparticles (LN) with the purpose of improving their toxicity profile. LN are made of lipids recognized as safe by the Food and Drug Administration (FDA) and, therefore, these systems are generally considered as nontoxic vehicles. However, toxicity studies regarding the use of LN as vehicles for drug administration are limited. In the present study, we investigated the in vivo toxicity of free edelfosine, and the protection conferred by LN. The free drug, non-loaded LN and edelfosine-loaded LN were orally administered to mice. Our results show that the oral administration of the free drug at 4 times higher than the therapeutic dose caused the death of the animals within 72h. Moreover, histopathology revealed gastrointestinal toxicity and an immunosuppressive effect. In contrast, LN showed a protective effect against edelfosine toxicity even at the higher dose and were completely safe. LN are, therefore, a safe vehicle for the administration of edelfosine by the oral route. The nanosystems developed could be further used for the administration of other drugs.


Subject(s)
Gastrointestinal Diseases/prevention & control , Lipids/pharmacology , Nanoparticles/chemistry , Phospholipid Ethers/antagonists & inhibitors , Phospholipid Ethers/toxicity , Administration, Oral , Animals , Dose-Response Relationship, Drug , Female , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/pathology , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/toxicity , Lipids/administration & dosage , Lipids/chemistry , Male , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Phospholipid Ethers/administration & dosage
18.
Biochemistry ; 53(23): 3719-26, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24832237

ABSTRACT

In the present study, we report the structure of the free and drug-bound Fab fragment of a high affinity anti-methotrexate antibody and perform a thermodynamic analysis of the binding process. The anti-methotrexate Fab fragment features a remarkably rigid tunnel-like binding site that extends into a water channel serving as a specialized route to move solvent out and into the site upon ligand binding and dissociation. This new finding in antibody structure-function relationships directly relates to the fast association (1 × 107 M⁻¹ s⁻¹) and slow dissociation (4 × 10⁻5 s⁻¹) rates determined for mAb ADD056, resulting in a very strong binding with a K(D) ~ 3.6 pM at 20 °C. As follows from the X-ray data analysis, the methotrexate-antibody complex is stabilized by an extended network of hydrogen bonds and stacking interactions. The analysis also shows structural involvement of the CDR H3 in formation of the water channel revealing another important role of this hypervariable region. This suggests a new direction in natural affinity maturation and opens a new possibility in antibody engineering. Methotrexate is a widely used therapeutic agent for many malignant diseases and inflammatory disorders. Unfortunately, it may also interfere with central aspects of metabolism and thereby cause inevitable side effects. Therefore, methotrexate therapy requires careful monitoring of drug blood levels, which is traditionally done by immunoassays. An understanding of the structure-function properties of antibodies selected for drug monitoring substantiates the performance and robustness of such tests.


Subject(s)
Antibodies, Monoclonal/metabolism , Binding Sites, Antibody , Folic Acid Antagonists/chemistry , Immunoglobulin Fab Fragments/metabolism , Methotrexate/antagonists & inhibitors , Models, Molecular , Water/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibody Affinity , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/metabolism , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , Crystallography, X-Ray , Folic Acid Antagonists/metabolism , Hybridomas , Hydrogen Bonding , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/metabolism , Ligands , Methotrexate/chemistry , Methotrexate/metabolism , Mice , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thermodynamics , Water/chemistry
19.
Hypertens Res ; 37(8): 724-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24718302

ABSTRACT

Chronic immunosuppressive therapy is often complicated by the development of both arterial hypertension and renal dysfunction. The principal aim of this study was to assess the effects of dual inhibition of renin-angiotensin system (RAS) and other antihypertensive treatment on blood pressure and renal function in normotensive and hypertensive Fawn-Hooded (FH) strains during chronic calcineurin inhibitor (CNI) administration. Combinations of perindopril (5 mg kg(-1) per day) and losartan (50 mg kg(-1) per day) or amlodipine (6 mg kg(-1) per day) and metoprolol (80 mg kg(-1) per day) were administered to normotensive (FHL) and hypertensive (FHH) rats, fed with diet containing tacrolimus (Tac; 12 mg kg(-1) per day). Tac-induced arterial hypertension in both animal strains (FHL: 151±4; FHH: 198±6 mm Hg) was prevented by dual RAS inhibition (FHL: 132±3 mm Hg, P<0.05; FHH: 153±3 mm Hg, P<0.05) as well as by a combination of amlodipine and metoprolol (FHL: 136±3 mm Hg, P<0.05; FHH: 166±4 mm Hg, P<0.05). However, significant nephroprotection was observed only in animals on dual RAS inhibition where albuminuria was reduced in both FHL (51.1±3.9 vs. 68.3±4.5 µg per day; P<0.05) and FHH rats (13.1±0.3 vs. 18.8±0.7 mg per day; P<0.05). We also found Tac-induced enhancement in renal angiotensin II activity that was significantly reduced by dual RAS inhibition in both FHL (63.5±3.2 vs. 23.1±3.0 fmol g(-1)) and FHH (79.8±8.5 vs. 32.2±5.8 fmol g(-1)). In addition, histological analysis revealed that RAS inhibition noticeably diminished glomerulosclerosis and tubulo-interstitial injury. This study indicates that dual blockade of RAS significantly attenuates Tac-induced arterial hypertension and nephrotoxicity in FH rats and further supports the notion that RAS inhibitors display efficient renoprotective properties during CNI treatment.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/toxicity , Renin-Angiotensin System/drug effects , Tacrolimus/antagonists & inhibitors , Tacrolimus/toxicity , Adrenergic beta-Antagonists/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Calcineurin/drug effects , Calcium Channel Blockers/pharmacology , Diet , Male , Rats
20.
Immunol Invest ; 43(6): 517-34, 2014.
Article in English | MEDLINE | ID: mdl-24295450

ABSTRACT

Although cancer immunotherapy, which is able to target specifically cancer cells without detrimental effects to normal cell functions, would serve as an ideal therapeutic modality, most of the randomized clinical trials of cancer immunotherapy have not demonstrated a meaningful survival benefit to cancer patients over preexisting therapeutic modalities. Due to the discrepancy between the impressive preclinical results and the limited clinical results, the cancer immunotherapy is not accepted generally as a standard therapy for cancers. A variety of immune escape mechanisms are thought to be involved in this ineffectiveness of cancer immunotherapy. Therefore, elimination of immunosuppressive activities in tumor microenvironment will enhance the effectiveness of cancer immunotherapy, which is currently focused on activation of tumor-specific immune responses. Since there are now increasing evidences showing that many cytotoxic anticancer drugs including targeted agents given in lower-than-therapeutic doses have not only the ability to eliminate tumor cells but can also block the immunosuppressive activities in tumor microenvironments and consequently favor the development of anticancer immune responses, clinically available drugs can be considered for their rapid application to cancer immunotherapies to enhance the efficacy of cancer immunotherapies with marginal effects on cancer treatment.


Subject(s)
Cancer Vaccines , Drug Therapy , Immunotherapy, Adoptive , Neoplasms/therapy , Animals , Antigens, Neoplasm/immunology , Combined Modality Therapy , Humans , Immunosuppressive Agents/antagonists & inhibitors , Neoplasms/immunology , Tumor Escape/drug effects , Tumor Escape/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL