Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Mol Immunol ; 142: 63-75, 2022 02.
Article in English | MEDLINE | ID: mdl-34965485

ABSTRACT

Severe acute pancreatitis (SAP) is complicated by systemic inflammatory response syndrome and multiple organ dysfunction, the disease will eventually result in death in almost half of the case. The spleen, as the largest immune organ adjacent to the pancreas, is prone to damage in SAP, thereby aggravating the damage of other organs and increasing mortality. However, to date, the research on the mechanism and treatment of spleen injury caused by SAP is still in its infancy. Herein, we investigated the mechanism of spleen injury, and explored the application potential of tuftsin for relieving spleen damage in SAP mice. Firstly, SAP mice model was constructed via the retrograde infusion of 3.5 % sodium taurocholate into the biliopancreatic duct. Then, we proved that the up-regulation of Toll-like receptor 4 (TLR4) in spleen would lead to the accumulation of reactive oxygen species (ROS) and mitochondrial dysfunction under SAP conditions. The splenic ROS and mitochondrial dysfunction could be improved by N-acetylcysteine (NAC) treatment or knocking out TLR4 in SAP mice. Meanwhile, we found that NAC treatment could also improve the autophagy of spleen tissue, suggesting that splenic ROS may affect impaired autophagy, causing the accumulation of damaged mitochondria, aggravating spleen damage. Furthermore, we verified the mechanism of spleen injury is caused by splenic ROS affecting PI3K/p-AKT/mTOR pathway-mediated autophagy. In addition, we detected the spleen injury caused by SAP could decrease the concentration of tuftsin in the serum of mice. Whereas, exogenous supplementation of tuftsin ameliorated the pathological damage, ROS accumulation, impaired autophagy, inflammation expression and apoptosis in damaged spleen. In summary, we verified the new mechanism of SAP-caused spleen damage that TLR4-induced ROS provoked mitophagy impairment and mitochondrial dysfunction in spleen via PI3K/p-AKT mTOR signaling, and the application potential of tuftsin in treating spleen injury, which might expand novel ideas and methods for the treatment of pancreatitis.


Subject(s)
Mitophagy/physiology , Pancreatitis/pathology , Reactive Oxygen Species/metabolism , Spleen/pathology , Toll-Like Receptor 4/metabolism , Acetylcysteine/pharmacology , Animals , Apoptosis/physiology , Immunologic Factors/therapeutic use , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mitochondria/pathology , Pancreas/pathology , Pancreatitis/chemically induced , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Spleen/injuries , TOR Serine-Threonine Kinases/metabolism , Taurocholic Acid/toxicity , Toll-Like Receptor 4/genetics , Tuftsin/therapeutic use
2.
J Neuroimmunol ; 337: 577070, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31683117

ABSTRACT

Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) which carries a significant burden of morbidity and mortality. Herein we examine the effects of acute treatment with tuftsin-phosphorylcholine (TPC), a novel immune-modulating helminth derived compound, on a murine model of MS. Experimental autoimmune encephalomyelitis (EAE) mice received acute treatment with TPC showed an improved clinical score and significantly less signs of inflammation and demyelination in CNS tissue compared with vehicle treated EAE mice. Our findings suggest that TPC may provide a beneficial clinical effect in EAE and may therefore have a potential value for ameliorating clinical manifestations and delaying disease progression in MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Inflammation Mediators/antagonists & inhibitors , Phosphorylcholine/analogs & derivatives , Tuftsin/therapeutic use , Animals , Drug Combinations , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Inflammation Mediators/immunology , Mice , Mice, Inbred C57BL , Phosphorylcholine/therapeutic use
3.
J Autoimmun ; 98: 113-121, 2019 03.
Article in English | MEDLINE | ID: mdl-30638709

ABSTRACT

Tuftsin-PhosphorylCholine (TPC) is a novel bi-specific molecule which links tuftsin and phosphorylcholine. TPC has shown immunomodulatory activities in experimental mouse models of autoimmune diseases. We studied herein the effects of TPC ex vivo on both peripheral blood mononuclear cells (PBMCs) and temporal artery biopsies (TABs) obtained from patients with giant cell arteritis (GCA) and age-matched disease controls. GCA is an immune-mediated disease affecting large vessels. Levels of 18 cytokines in supernatants, PBMC viability, T helper (Th) cell differentiation of PBMCs and gene expression in TABs were analyzed. Treatment ex vivo with TPC decreased the production of IL-1ß, IL-2, IL-5, IL-6, IL-9, IL-12(p70), IL-13, IL-17A, IL-18, IL-21, IL-22, IL-23, IFNγ, TNFα, GM-CSF by CD3/CD28 activated PBMCs whereas it negligibly affected cell viability. It reduced Th1 and Th17 differentiation while did not impact Th22 differentiation in PBMCs stimulated by phorbol 12-myristate 13-acetate plus ionomycin. In inflamed TABs, treatment with TPC down-regulated the production of IL-1ß, IL-6, IL-13, IL-17A and CD68 gene expression. The effects of TPC were comparable to the effects of dexamethasone, included as the standard of care, with the exception of a greater reduction of IL-2, IL-18, IFNγ in CD3/CD28 activated PBMCs and CD68 gene in inflamed TABs. In conclusion our results warrant further investigations regarding TPC as an immunotherapeutic agent in GCA and potentially other autoimmune and inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Dexamethasone/therapeutic use , Giant Cell Arteritis/drug therapy , Immunotherapy/methods , Phosphorylcholine/analogs & derivatives , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Tuftsin/therapeutic use , Aged , Aged, 80 and over , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Drug Combinations , Female , Humans , Lymphocyte Activation , Male , Phosphorylcholine/therapeutic use
4.
Immunol Res ; 66(6): 637-641, 2018 12.
Article in English | MEDLINE | ID: mdl-30554380

ABSTRACT

The distinction that in areas where helminthic infections are common, autoimmune diseases are less prevalent, led to the investigation of immune modulatory properties of helminths and their derivatives. Such are phosphorylcholine (PC) moieties which are a component of secreted products of helminths. PC has been broadly studied for its attenuating effects on the human immune system. In an attempt to develop a novel therapeutic small molecule for the treatment of autoimmune conditions, we have conjugated PC with tuftsin, a natural immunomodulatory tetrapeptide, to create TPC. Herein, we review our findings regarding the effects of TPC in murine models of three autoimmune diseases-systemic lupus erythematosus (SLE), inflammatory bowel disease (IBD), and rheumatic arthritis (RA), as well as ex-vivo samples from giant cell arteritis (GCA) patients. In all four disease models examined, TPC was shown to attenuate the inflammatory response by reducing expression of pro-inflammatory cytokines and altering the phenotype of T cell expression. In murine models, TPC has further produced a significant improvement in clinical disease scores with no significant side effects noted. Our findings suggest TPC presents promising potential as a novel therapeutic agent for the effective treatment of various autoimmune conditions.


Subject(s)
Autoimmunity/drug effects , Helminths/drug effects , Phosphorylcholine/pharmacology , Phosphorylcholine/therapeutic use , Tuftsin/pharmacology , Tuftsin/therapeutic use , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/parasitology , Helminthiasis/drug therapy , Helminthiasis/parasitology , Humans , Inflammation/drug therapy , Inflammation/parasitology
5.
PLoS One ; 13(8): e0200615, 2018.
Article in English | MEDLINE | ID: mdl-30089122

ABSTRACT

A novel small molecule named tuftsin-phosphorylcholine (TPC), which is linked to the biological activity of helminths, was constructed. The current study address the effect of TPC treatment in established collagen-induced arthritis (CIA) mice and propose TPC bi-functional activity. TPC treatment was initiated when clinical score was 2 to 4. Arthritis scores in TPC treated mice were lower compared to mice treated with vehicle (P < 0.001). Joint staining showed normal joint structure in TPC-treated mice compared to control groups treated with phosphate buffered saline (PBS), phosphorylcholine, or tuftsin, which exhibited severely inflamed joints. TPC enhanced anti-inflammatory response due to increased IL-10 secretion, and reduced pro-inflammatory cytokine secretion (IL-1-ß, IL-6, TNF-αP < 0.001). Furthermore, TPC therapy increased expansion of CD4+CD25+FOXP3+T regulatory cells and IL-10+CD5+CD1d+B regulatory cells. We propose that the immunomodulatory activity of TPC can be a result of a bi-specific activity of TPC: (a) The tuftsin part of the TPC shifts RAW macrophage cells from pro-inflammatory macrophages M1 to anti-inflammatory M2-secreting IL-10 (P < 0.001) through neuropilin-1 and (b) TPC significantly reduce mouse TLR4 expression via NFkB pathway by HEKTM cells (P < 0.02) via the phosphorylcholine site of the molecule. Our results indicate that TPC, significantly ameliorated established CIA by its immunomodulatory activity. These data could lead to a novel self bi-functional small molecule for treating patients with progressive RA.


Subject(s)
Arthritis, Experimental/drug therapy , Helminths/metabolism , Phosphorylcholine/therapeutic use , Tuftsin/therapeutic use , Animals , Arthritis, Experimental/pathology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Joints/pathology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred DBA , NF-kappa B/metabolism , Neuropilin-1/metabolism , Phosphorylcholine/pharmacology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tuftsin/pharmacology
6.
Clin Exp Immunol ; 193(2): 160-166, 2018 08.
Article in English | MEDLINE | ID: mdl-29698559

ABSTRACT

The role of helminth treatment in autoimmune diseases is growing constantly. Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease with challenging treatment options. Tuftsin-phosphorylcholine (TPC) is a novel helminth-based compound that modulates the host immune network. This study was conducted to evaluate the potential value of TPC in ameliorating lupus nephritis in a murine model and specifically to compare the efficacy of TPC to the existing first-line therapy for SLE: corticosteroids (methylprednisolone). Lupus-prone NZBxW/F1 mice were treated with TPC (5 µg/mouse), methylprednisolone (MP; 5 mg/body weight) or phosphate-buffered saline (PBS) (control) three times per week once glomerulonephritis, defined as proteinuria of grade > 100 mg/dl, was established. Levels of anti-dsDNA autoantibodies were evaluated by enzyme-linked immunosorbent assay (ELISA), splenic cytokines were measured in vitro and the kidney microscopy was analysed following staining. TPC and MP treatments improved lupus nephritis significantly and prolonged survival in NZBxW/F1 mice. TPC-treated mice showed a significantly decreased level of proteinuria (P < 0·001) and anti-dsDNA antibodies (P < 0·001) compared to PBS-treated mice. Moreover, TPC and MP inhibited the production of the proinflammatory cytokines interferon IFN-γ, interleukin IL-1ß and IL-6 (P < 0·001) and enhanced expression of the anti-inflammatory cytokine IL-10 (P < 0·001). Finally, microscopy analysis of the kidneys demonstrated that TPC-treated mice maintained normal structure equally to MP-treated mice. These data indicate that the small molecule named TPC hinders lupus development in genetically lupus-prone mice equally to methylprednisolone in most of the cases. Hence, TCP may be employed as a therapeutic potential for lupus nephritis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Helminths/immunology , Kidney/pathology , Lupus Erythematosus, Systemic/drug therapy , Lupus Nephritis/drug therapy , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/therapeutic use , Tuftsin/therapeutic use , Animals , Antibodies, Antinuclear/blood , Cytokines/metabolism , Disease Models, Animal , Drug Combinations , Female , Humans , Inflammation Mediators/metabolism , Kidney/drug effects , Methylprednisolone/therapeutic use , Mice , Mice, Inbred NZB , Phosphorylcholine/chemistry , Tuftsin/chemistry
7.
Curr Med Chem ; 24(34): 3711-3727, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-28745220

ABSTRACT

BACKGROUND: Immunomodulation is one of the significant therapeutic strategies. It includes both stimulation and suppression of the immune system by a variety of substances called immunomodulators, designed to regulate the immune response of the organism against infections of varying etiology. An example of such a substance is tuftsin (TKPA) 3 (Fig. (1)). In this paper were included tuftsin derivatives, which were described over the years, their together with biological activity and clinical potential. METHODS: We reviewed a bibliographic database to gather all the important information about the tuftsin peptide. We have delineated the significant information on the activity of the tetrapeptide itself and its derivatives. Analogs were divided because of their anti-tumor, anti-inflammatory, antimicrobial and anti-viral activity. RESULTS: This paper describes eighty-six documents. Thirty-two of them concern on activity of tuftsin in the human organism. The remaining fifty-four describe peptide analogues and their properties, including eleven papers about the tuftsin-based peptides contained in the vaccines, nine papers representing anticancer activity of the tuftsin derivatives, twenty-six about antiinflammatory compounds, and five papers describing the antitumor activity of the tuftsin analogs. CONCLUSION: The findings of this review confirm the importance of the tuftsin and their derivatives. Most of these substances showed anti-tumor, anti-inflammatory or antibacterial activities. A large amount of the compounds may find use in vaccines. Tuftsin can also be used to prepare fusion proteins in the treatment of cancer and as carriers of many biologically active substances.


Subject(s)
Tuftsin/analogs & derivatives , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Cytokines/metabolism , Fullerenes/chemistry , Humans , Mycobacterium tuberculosis/drug effects , Neoplasms/drug therapy , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tuftsin/therapeutic use
8.
Anticancer Drugs ; 25(8): 857-67, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24714081

ABSTRACT

Immune adjuvants have been used in cancer biotherapies to stimulate immune response to tumor cells. Despite their potential as anticancer reagents, there are several impediments to their use in clinical applications. In this study, we aim to modify the existing tuftsin structure and evaluate its antitumor activity in preclinical models. We synthesized a novel tuftsin derivative, namely, the T peptide (TP), by linking four tuftsin peptides, which showed enhanced stability in vivo. We then evaluated its anticancer activity in a postoperative residual tumor model in mice, where we surgically removed most of the primary tumor from the host, a procedure mimicking clinically postoperative patients. Despite the limited effect in intact solid tumors, TP strongly inhibited relapsed growth of residual tumors in postsurgical mice. Surgical resection of tumors accelerated residual tumor growth, but TP slowed down this process significantly. Interestingly, TP showed similar effects in human xenograft residual models. As an immunomodulator, TP could synergize the functions of macrophages, thus inhibiting the growth of cocultured tumor cells in vitro. Furthermore, TP could shift the macrophages to the tumor-suppressive M1 type and mobilize them to produce elevated cytotoxic TNF-α and NO. As a result, TP effectively prolonged the survival time of tumor-resected mice. Using the postoperative residual tumor models, we provide a body of evidence showing the antitumor activity of TP, which causes no obvious toxicity. Our study highlights the potential of TP as a postoperative adjuvant in cancer therapies.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasm, Residual/drug therapy , Tuftsin/analogs & derivatives , Tuftsin/therapeutic use , Adjuvants, Immunologic/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Disease-Free Survival , Humans , Lysine/chemistry , Macrophages/drug effects , Macrophages/immunology , Mice , Neoplasm, Residual/immunology , Neoplasm, Residual/pathology , Tuftsin/chemistry , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
9.
Vaccine ; 27(3): 369-74, 2009 Jan 14.
Article in English | MEDLINE | ID: mdl-19027810

ABSTRACT

Pharmacological manipulation of the balance between pro- and anti-inflammatory mediators emerges as a key aspect of a successful treatment of sepsis. A murine model of septic shock was developed and chosen conjugates (1a, 1b, 8a, 8c) and analogs (T2) of muramyl dipeptide and tuftsin were tested in this model as prospective anti-bacterial drugs or adjuvants. The phagocytic activity of monocytes/macrophages was determined (flow cytometry, bacterial clearance from vital organs). To evaluate cytokines levels (TNFalpha, IFNgamma, IL6, IL10) we used real-time PCR. The most promising immunomodulatory properties were displayed by the analogue T2 and two conjugates: 8a, 8c.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/therapeutic use , Immunologic Factors/therapeutic use , Inflammation/drug therapy , Sepsis/drug therapy , Tuftsin/therapeutic use , Animals , Colony Count, Microbial , Cytokines/biosynthesis , Escherichia coli Infections/drug therapy , Gene Expression Profiling , Liver/microbiology , Lung/microbiology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Phagocytosis , Polymerase Chain Reaction/methods
10.
Pharmacol Rep ; 58(4): 562-7, 2006.
Article in English | MEDLINE | ID: mdl-16963804

ABSTRACT

TP-7 is a synthetic analogue of tuftsin. It has a structure of tuftsin, to which three natural L-amino-acids Pro-Gly-Pro are attached. This heptapeptide improves learning and memorization and causes antidepressant and anxiolytic effect. It is possible to use TP-7 in the future to optimize cognitive functions and as a potential new anxioselective, fast-acting and easy-dosed drug. Therefore, it was purposeful to study such properties of the heptapeptide as its influence on anxiety-fear and body weight under a long-term treatment regimen. The experiment was performed on 24 preselected Wistar rats with the use of Rodina's method. There were three experimental groups of animals with high initial emotional reactivity: passive control group (P), active control group (A, receiving distilled water) and group treated with TP-7 at the dose of 0.3 mg/kg (T). The rats of A and T groups received intraperitoneal injections every day. The experiments were conducted 15 min after the administration of the drug, one and two days after initial testing day, then 1, 2, 3 and 4 weeks after that. The heptapeptide reduced the anxiety-phobic states significantly starting from the second day of drug application. The observed effects persisted throughout four weeks of the experiment, which confirmed effective long-term anxiolytic properties of the heptapeptide. TP-7 did not cause any changes in the body mass by itself.


Subject(s)
Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Body Weight/drug effects , Oligopeptides/pharmacology , Tuftsin/pharmacology , Animals , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Fear/drug effects , Male , Motor Activity/drug effects , Oligopeptides/therapeutic use , Phobic Disorders/drug therapy , Rats , Rats, Wistar , Reaction Time/drug effects , Time Factors , Tuftsin/analogs & derivatives , Tuftsin/therapeutic use
11.
J Drug Target ; 14(4): 233-41, 2006 May.
Article in English | MEDLINE | ID: mdl-16777682

ABSTRACT

Present study was performed to evaluate the efficacy, toxicity and pharmacokinetics of antifungal drug nystatin incorporated in immunomodulator tuftsin-bearing liposomes. In vitro toxicity of free nystatin and nystatin incorporated in tuftsin-free or tuftsin-loaded liposomes was assessed by incubation of nystatin formulations with human erythrocytes. The toxicity profile of free nystatin and liposomal formulations of nystatin with or without tuftsin was also analyzed by monitoring the level of blood urea nitrogen (BUN) and serum creatinine in the treated BALB/c mice. The results of the present work showed that tuftsin-loaded nystatin liposomes like conventional nystatin liposomes exerted less toxicity to human erythrocytes as compared with free nystatin. Moreover, mice treated with tuftsin-loaded nystatin liposomes showed insignificant elevation in the biochemical values of serum creatinine and blood urea. The stability of nystatin liposomes upon incorporation of tuftsin was evaluated by monitoring the leakage of the entrapped drug in human serum. Tuftsin-loaded liposomes held nystatin for longer duration in the presence of serum than identical nystatin liposomes without tuftsin. Pharmacokinetics of the both tuftsin-free or tuftsin-loaded liposomal formulations nystatin was analyzed by determining the level of nystatin in the systemic circulation of mice at different time points. Mice injected with tuftsin-loaded nystatin liposomes showed higher level of the drug in the systemic circulation compared with those treated with conventional nystatin liposomes. The efficacy of tuftsin-loaded nystatin liposomes against A. fumigatus was evaluated by assessing the fungal burden in the lungs of treated mice. Treatment with tuftsin-loaded nystatin liposomes was most effective in eliminating fungal burden from lung tissues of infected mice compared to those treated with free nystatin or nystatin liposomes without tuftsin. The immunopotentiating activity, increased stability and less toxicity of tuftsin-incorporated nystatin liposomes, supports the idea for its prophylactic and therapeutic use in the clinical setting.


Subject(s)
Antifungal Agents , Aspergillosis/drug therapy , Immunologic Factors , Liposomes , Nystatin , Tuftsin , Animals , Antifungal Agents/adverse effects , Antifungal Agents/pharmacokinetics , Antifungal Agents/therapeutic use , Aspergillosis/microbiology , Aspergillus fumigatus/drug effects , Disease Models, Animal , Humans , Immunologic Factors/adverse effects , Immunologic Factors/chemistry , Immunologic Factors/pharmacokinetics , Immunologic Factors/therapeutic use , Liposomes/chemistry , Male , Mice , Microbial Sensitivity Tests , Nystatin/adverse effects , Nystatin/chemistry , Nystatin/pharmacokinetics , Nystatin/therapeutic use , Treatment Outcome , Tuftsin/adverse effects , Tuftsin/chemistry , Tuftsin/pharmacokinetics , Tuftsin/therapeutic use
12.
J Antimicrob Chemother ; 58(1): 125-32, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16709592

ABSTRACT

OBJECTIVES: In the present study we evaluated the pharmacokinetics and toxicity of amphotericin B in immunomodulator tuftsin-loaded liposomes in a murine model. METHODS: Stability of amphotericin B liposomes was tested by incubating one volume of liposomal formulations of amphotericin B with nine volumes of serum. The pharmacokinetics of amphotericin B in Candida albicans-infected mice treated with conventional and tuftsin-loaded amphotericin B liposomes was evaluated over a period of 24 h. In vitro toxicity of amphotericin B deoxycholate, as well as amphotericin B liposomes, was tested by incubation with human erythrocytes for 1 h at 37 degrees C. To assess amphotericin B-induced in vivo toxicity, BALB/c mice were injected with three doses of amphotericin B deoxycholate, as well as amphotericin B liposomal formulations on days 1, 2 and 3 post C. albicans infection. Blood from treated mice was taken by retro-orbital puncture to test renal function parameters such as serum creatinine and urea. RESULTS: In vitro stability studies revealed that tuftsin-bearing amphotericin B liposomes released only 11% of the total liposomal amphotericin B in the serum, while it was found to be 19% from identical tuftsin-free amphotericin B liposomes. Both tuftsin-loaded as well as tuftsin-free liposomal formulations of amphotericin B induced approximately 20% haemolysis of erythrocytes at a dose of 40 mg/L, while the same amount of drug in amphotericin B deoxycholate caused 100% lysis of the erythrocytes. Pharmacokinetic studies revealed that subsequent to administration of various formulations of amphotericin B, there was 32 mg/L amphotericin B in the systemic circulation of mice treated with tuftsin-bearing amphotericin B liposomes, while it was 25 mg/L for amphotericin B liposomes, 4 h post drug administration. In vivo toxicity studies demonstrated that the amphotericin B deoxycholate formulation induced elevations in serum creatinine (approximately 300% of control) and blood urea (approximately 380% of control) values, while these values were substantially less (blood urea approximately 150% of control and serum creatinine approximately 210% of control) in the animals treated with the tuftsin-loaded amphotericin B liposomal formulation. Further, the administration of amphotericin B deoxycholate (1 mg/kg) in BALB/c mice at a dose of 1 mg/kg body weight led to the accumulation of 18.6 +/- 5.25 g/kg (of amphotericin B) in kidneys. On the other hand, administration of liposomal amphotericin B and tuftsin-bearing liposomal amphotericin B at a dose of 5 mg/kg body weight resulted in accumulation of 8.8 +/- 2.0 and 4.0 +/- 1.6 g/kg of amphotericin B, respectively, in the kidneys of treated animals. CONCLUSIONS: Co-administration of immunomodulator tuftsin along with liposomal formulations of amphotericin B successfully minimizes toxicity, as well as other side effects of the drug. Interestingly, tuftsin also increased the stability of liposomal amphotericin B. Superior efficacy, reliable safety and favourable pharmacodynamics of tuftsin-loaded amphotericin B liposomes suggest their potential therapeutic value in the management of fungal infections.


Subject(s)
Amphotericin B/adverse effects , Amphotericin B/pharmacokinetics , Liposomes/chemistry , Tuftsin/adverse effects , Tuftsin/pharmacokinetics , Amphotericin B/chemistry , Amphotericin B/therapeutic use , Animals , Antifungal Agents/adverse effects , Antifungal Agents/chemistry , Antifungal Agents/pharmacokinetics , Antifungal Agents/therapeutic use , Candidiasis/drug therapy , Deoxycholic Acid/adverse effects , Deoxycholic Acid/pharmacokinetics , Deoxycholic Acid/therapeutic use , Dose-Response Relationship, Drug , Drug Combinations , Drug Stability , Immunologic Factors/adverse effects , Immunologic Factors/chemistry , Immunologic Factors/pharmacokinetics , Immunologic Factors/therapeutic use , Mice , Mice, Inbred BALB C , Tuftsin/chemistry , Tuftsin/therapeutic use
13.
Acta Pol Pharm ; 63(5): 378-81, 2006.
Article in English | MEDLINE | ID: mdl-17357588

ABSTRACT

Phagocytosis plays a crucial role in a host defense against invading microorganisms. This process can be induced by many phagocytosis stimulating factors. One of them is an endogenous tetrapeptide - tuftsin that occurs in the blood of mammals including human beings. Tuftsin is capable of potentiating granulocyte and macrophage functions such as: phagocytosis, motility, and chemotaxis as well as bactericidal and tumoricidal activity. The other particle able to induce phagocytosis is muramyl dipeptide (MDP), the smallest synthetic glycopeptide of bacterial origin that possesses an immunogenic activity. MDP is known to affect most functions of macrophages. Phagocytosis stimulating properties of a new group of tuftsin and MDP analogues (one tuftsin analogue and four conjugates of tuftsin/retro tuftsin and muramyl dipeptide or nor-muramyl dipeptide) were tested. The results of the study show that all of the examined conjugates are able to generate oxidative burst. The most promising analogues proved to be kd6 and kd7.


Subject(s)
Immunotherapy/methods , Phagocytosis/drug effects , Tuftsin/therapeutic use , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Adjuvants, Immunologic , Cells, Cultured , Drug Evaluation, Preclinical , Granulocytes/cytology , Granulocytes/drug effects , Humans , Monocytes/cytology , Monocytes/drug effects , Respiratory Burst/drug effects , Tuftsin/analogs & derivatives
14.
J Drug Target ; 13(7): 423-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16308211

ABSTRACT

The co-administration of immunomodulators and antibiotics has been proved very successful for treatment of opportunistic infectious diseases. In the present study, we evaluated the combination of liposomal amphotericin B (lip-Amp B) and immunomodulator tuftsin to cure Cryptococcus neoformans infection in BALB/c mice. Mice infected with C. neoformans were treated with Amp B deoxycholate and tuftsin free or tuftsin-loaded Amp B liposomes. The results of the present study demonstrated higher efficacy of tuftsin-loaded Amp B liposomes against experimental murine cryptococcosis, in terms of enhanced survival rate and reduced fungal burden in organs (lungs and brain) of the treated mice. Interestingly, pre-treatment of mice with liposomal tuftsin before challenging them with the C. neoformans infection resulted in 100% survival of the treated animals followed by treatment with lip-Amp B. Immunomodulator-based therapy seems likely to be more beneficial for treatment of fungal infectious diseases.


Subject(s)
Amphotericin B/therapeutic use , Antifungal Agents/therapeutic use , Cryptococcosis/drug therapy , Cryptococcus neoformans/drug effects , Immunologic Factors/therapeutic use , Tuftsin/therapeutic use , Amphotericin B/administration & dosage , Animals , Antifungal Agents/administration & dosage , Cryptococcosis/microbiology , Drug Therapy, Combination , Female , Immunocompetence , Immunologic Factors/administration & dosage , Liposomes , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Tuftsin/administration & dosage
15.
FEMS Immunol Med Microbiol ; 44(3): 269-76, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15907448

ABSTRACT

In the present study, we evaluated the immunopotentiating efficacy of tuftsin against experimental murine aspergillosis in both normal and immunodebilitant BALB/c mice. The animals were challenged with an isolate of Aspergillus fumigatus (1x10(8) cfu/mouse) that was showing less susceptibility to lower doses of amphotericin B in murine animal model. Co-administration of the immunomodulator tuftsin and liposomised-amphotericin B was found to be highly effective in the treatment of systemic infection of A. fumigatus in both immunocompetent and leukopenic mice. Moreover, pre-treatment of mice with liposomised-tuftsin prior to challenging them with A. fumigatus infection and subsequent treatment with tuftsin-bearing liposomised-amphotericin B was found to be extremely efficient in successful elimination of fungal pathogen. In another set of experiments, tuftsin-mediated antigen-specific memory antibody response was also assessed by immunizing the animals with A. fumigatus cytosolic antigen. The animals that received a booster 150 days after the first immunization with tuftsin-liposomes-antigen showed more resistance to A. fumigatus infection in comparison with the naïve animals.


Subject(s)
Amphotericin B/therapeutic use , Antifungal Agents/therapeutic use , Aspergillosis/immunology , Aspergillosis/prevention & control , Aspergillus fumigatus , Immunologic Factors/therapeutic use , Tuftsin/therapeutic use , Amphotericin B/pharmacology , Animals , Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Drug Evaluation, Preclinical , Drug Therapy, Combination , Immunocompromised Host , Immunologic Factors/pharmacology , Liposomes , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests
16.
Stroke ; 36(3): 613-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15692122

ABSTRACT

BACKGROUND AND PURPOSE: Microglial activation may contribute to the pathogenesis of the brain injury in intracerebral hemorrhage (ICH). We have reported that the tripeptide macrophage/microglial inhibitory factor (MIF), Thr-Lys-Pro, inhibits microglial activation and results in functional improvement when given before the onset of hemorrhage. In this study, we investigate the protection and efficacy of treatment when MIF is administered 2 hours after collagenase injection. METHODS: ICH was induced by injecting bacterial collagenase into the caudate nucleus; 100 microL MIF (500 micromol/L) was delivered via a micro-osmotic pump. Infusion of MIF or saline (control) was initiated 2 hours after collagenase injection and continued for 24 or 72 hours. Microglial activation and macrophage infiltration were assessed by 5-d-4 and F4/80 immunofluorescence, respectively. Production of reactive oxygen species was visualized by in situ detection of ethidium. Degenerating neurons were assessed by Fluoro-Jade B staining. Neurological deficits, brain injury volumes, and brain edema were assessed at 24 and 72 hours after MIF/saline treatment. RESULTS: MIF can inhibit microglial activation and macrophage infiltration, attenuate the numbers of ethidium-positive cells compared with the saline-treated control mice, reduce the injury volume, edema, and degenerating neurons, and improve the neurological functional outcome. CONCLUSIONS: Activated microglia/macrophages are important contributors to brain injury after ICH. MIF could be a valuable neuroprotective agent for the treatment of ICH, if treatment is initiated soon after the onset of hemorrhage.


Subject(s)
Brain Injuries/prevention & control , Cerebral Hemorrhage/chemically induced , Peptide Fragments/therapeutic use , Tuftsin/therapeutic use , Animals , Caudate Nucleus/pathology , Cell Death/drug effects , Cerebral Hemorrhage/complications , Collagenases/adverse effects , Edema/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/physiology , Macrophage Migration-Inhibitory Factors/therapeutic use , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/physiology , Neurons/drug effects , Psychomotor Disorders/etiology , Psychomotor Disorders/prevention & control , Reactive Oxygen Species/metabolism , Stroke Volume/drug effects
17.
Methods Enzymol ; 391: 291-304, 2005.
Article in English | MEDLINE | ID: mdl-15721387

ABSTRACT

Tuftsin is a tetrapeptide (Thr-Lys-Pro-Arg) that specifically binds monocytes, macrophages, and polymorphonuclear leukocytes and potentiates their natural killer activity against tumors and pathogens. The antimicrobial activity of this peptide is significantly increased by attaching at the C-terminus a fatty acyl residue through the ethylenediamine spacer arm. This activity is further augmented by incorporating the modified tuftsin in the liposomes. The tuftsin-bearing liposomes not only enhance the host's resistance against a variety of infections but also serve as useful vehicles for the site-specific delivery of drugs in a variety of macrophage-based infections, such as tuberculosis and leishmaniasis.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Drug Carriers , Liposomes , Macrophages/microbiology , Tuftsin/administration & dosage , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Aspergillosis/drug therapy , Erythrocytes/chemistry , Erythrocytes/metabolism , Leishmania donovani/metabolism , Leishmaniasis, Visceral/drug therapy , Liposomes/chemical synthesis , Liposomes/chemistry , Macrophages/cytology , Mice , Mycobacterium tuberculosis/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Rats , Tuberculosis/drug therapy , Tuftsin/chemistry , Tuftsin/metabolism , Tuftsin/therapeutic use
18.
Bull Exp Biol Med ; 138(2): 163-4, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15662460

ABSTRACT

Taftsin, an endogenous peptide immunostimulator, exhibits also high antiulcer activity. In a dose of 0.6 micromol/kg Taftsin decreases the area of ulcerative lesions in albino rats on various models of ulceration.


Subject(s)
Stomach Ulcer/drug therapy , Tuftsin/therapeutic use , Acetic Acid , Adjuvants, Immunologic/therapeutic use , Animals , Ethanol , Male , Rats , Restraint, Physical , Stomach Ulcer/chemically induced , Stress, Physiological/physiopathology
19.
J Drug Target ; 11(4): 247-51, 2003 May.
Article in English | MEDLINE | ID: mdl-14578113

ABSTRACT

In the present study, we evaluated the potential of an immunomodulator tuftsin in increasing the efficacy of liposomised diethylcarbamazine (DEC) against experimental filarial infection of Brugia malayi. The liposomised form of DEC, when used at sub-optimal dose of 25 mg/kg body weight, successfully eliminated filarial parasite from systemic circulation in animals inflicted with B. malayi infection. However, the formulation was effective upto 60 days post infection only, followed by recurrence of the infection. In contrast, the co-administration of liposomal formulation of DEC along with an immunomodulator tuftsin was found to be competent enough to suppress microfilarial stage of parasite till 90 days post treatment. Interestingly, tuftsin bearing DEC liposomes were found to be effective against adult parasite as well.


Subject(s)
Adjuvants, Immunologic/pharmacology , Brugia malayi/drug effects , Diethylcarbamazine/pharmacology , Filariasis/drug therapy , Tuftsin/pharmacology , Adjuvants, Immunologic/therapeutic use , Animals , Culicidae/parasitology , Diethylcarbamazine/therapeutic use , Drug Synergism , Female , Filariasis/parasitology , Filaricides/pharmacology , Filaricides/therapeutic use , Male , Muridae , Tuftsin/therapeutic use
20.
J Drug Target ; 11(2): 93-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12881195

ABSTRACT

In the present study, we evaluated tuftsin bearing nystatin liposomes for their potential against an isolate of Candida albicans (C. albicans) showing less in vivo susceptibility to amphotericin B (Amp B). The liposomised-Amp B in higher doses was found to be effective in elimination of less susceptible strain of C. albicans (C. albicans JMCR) in Balb/c mice, but may not be recommended due to toxicity constraints. On the other hand, liposomal nystatin was shown to possess higher efficacy as compared to that of Amp B, and was pertinent in treatment of C. albicans JMCR strain. The data of present work reveals that the incorporation of nystatin in tuftsin-bearing-liposomes results in a significant increase in its efficacy against experimental murine candidiasis. Interestingly, the pre-treatment of animals with liposomised-tuftsin prior to challenge with C. albicans infection was more effective in elimination of the pathogen from host and shows an advantage in prophylactic perspectives.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Amphotericin B/administration & dosage , Antifungal Agents/administration & dosage , Candidiasis/drug therapy , Nystatin/administration & dosage , Tuftsin/administration & dosage , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/therapeutic use , Amphotericin B/chemistry , Amphotericin B/therapeutic use , Animals , Antibiotic Prophylaxis , Antifungal Agents/chemistry , Antifungal Agents/therapeutic use , Candida albicans/drug effects , Candidiasis/prevention & control , Cholesterol , Chromatography, High Pressure Liquid , Female , Liposomes , Mice , Mice, Inbred BALB C , Nystatin/chemistry , Nystatin/therapeutic use , Phosphatidylcholines , Tuftsin/chemistry , Tuftsin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...