Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Gen Physiol Biophys ; 43(5): 457-467, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39140686

ABSTRACT

In this study, we investigated the effects of peripheral nesfatin-1 on basal brain activity and 4-aminopyridine (4-AP)-induced epileptiform activity, and its relationship with the electrocorticogram (ECoG) power spectrum and EEG bands. Forty-nine male Wistar rats were divided into seven groups: control sham, 4-AP (2.5 mg/kg i.p.), Nesfatin-1 (1, 2, and 4 µg/kg i.p.), Nesfatin-1 (2 µg/kg) post-treatment, and Nesfatin-1 (2 µg/kg) pre-treatment. Recordings were conducted for 70 min under ketamine/xylazine (90/10 mg/kg) anesthesia. In the post-treatment group, nesfatin-1 was injected 20 min after 4-AP induction. In the pre-treatment groups, nesfatin-1 was administered following basal recordings and before 4-AP injection. 4-AP induced epileptiform activity in all animals, peaking at 30 min. Nesfatin-1 (2 µg/kg) reduced basal brain activity (p < 0.05) and decreased alpha, delta, and theta bands in ECoG. Post-treatment of nesfatin-1 did not affect 4-AP-induced activity (p > 0.05) but increased gamma band activity (p > 0.05). Pre-treatment of nesfatin-1 reduced epileptiform activity between 50 and 60 min (p < 0.05), decreased delta bands, and increased gamma bands (p > 0.05). We conclude that peripheral nesfatin-1 modulates normal brain activity but has limited effects on abnormal discharges.


Subject(s)
Brain , Epilepsy , Nucleobindins , Rats, Wistar , Animals , Male , Rats , Epilepsy/physiopathology , Epilepsy/chemically induced , Epilepsy/blood , Brain/drug effects , Brain/metabolism , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/administration & dosage , Electroencephalography , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Treatment Outcome , Anticonvulsants/pharmacology , Anticonvulsants/administration & dosage
2.
Biochem Pharmacol ; 224: 116220, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641307

ABSTRACT

Alpha-enolase (ENO1), a multifunctional protein with carcinogenic properties, has emerged as a promising cancer biomarker because of its differential expression in cancer and normal cells. On the basis of this characteristic, we designed a cell-targeting peptide that specifically targets ENO1 and connected it with the drug doxorubicin (DOX) by aldehyde-amine condensation. A surface plasmon resonance (SPR) assay showed that the affinity for ENO1 was stronger (KD = 2.5 µM) for the resulting cell-targeting drug, DOX-P, than for DOX. Moreover, DOX-P exhibited acid-responsive capabilities, enabling precise release at the tumor site under the guidance of the homing peptide and alleviating DOX-induced cardiotoxicity. An efficacy experiment confirmed that, the targeting ability of DOX-P toward ENO1 demonstrated superior antitumor activity against colorectal cancer than that of DOX, while reducing its toxicity to cardiomyocytes. Furthermore, in vivo metabolic distribution results indicated low accumulation of DOX-P in nontumor sites, further validating its targeting ability. These results showed that the ENO1-targeted DOX-P peptide has great potential for application in targeted drug-delivery systems for colorectal cancer therapy.


Subject(s)
Antibiotics, Antineoplastic , Colorectal Neoplasms , Doxorubicin , Drug Delivery Systems , Phosphopyruvate Hydratase , Tumor Suppressor Proteins , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Phosphopyruvate Hydratase/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Animals , Tumor Suppressor Proteins/metabolism , Humans , Mice , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Drug Delivery Systems/methods , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/administration & dosage , Mice, Inbred BALB C , Mice, Nude , Male , Cell Line, Tumor , HCT116 Cells , Xenograft Model Antitumor Assays/methods , Biomarkers, Tumor
3.
Sci Rep ; 11(1): 22745, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815492

ABSTRACT

Although Krüppel-associated box domain-containing zinc-finger proteins (K-ZNFs) may be associated with sophisticated gene regulation in higher organisms, the physiological functions of most K-ZNFs remain unknown. The Zfp212 protein was highly conserved in mammals and abundant in the brain; it was mainly expressed in the cerebellum (Cb). Zfp212 (mouse homolog of human ZNF212) knockout (Zfp212-KO) mice showed a reduction in survival rate compared to wild-type mice after 20 months of age. GABAergic Purkinje cell degeneration in the Cb and aberrant locomotion were observed in adult Zfp212-KO mice. To identify genes related to the ataxia-like phenotype of Zfp212-KO mice, 39 ataxia-associated genes in the Cb were monitored. Substantial alterations in the expression of ataxin 10, protein phosphatase 2 regulatory subunit beta, protein kinase C gamma, and phospholipase D3 (Pld3) were observed. Among them, Pld3 alone was tightly regulated by Flag-tagged ZNF212 overexpression or Zfp212 knockdown in the HT22 cell line. The Cyclic Amplification and Selection of Targets assay identified the TATTTC sequence as a recognition motif of ZNF212, and these motifs occurred in both human and mouse PLD3 gene promoters. Adeno-associated virus-mediated introduction of human ZNF212 into the Cb of 3-week-old Zfp212-KO mice prevented Purkinje cell death and motor behavioral deficits. We confirmed the reduction of Zfp212 and Pld3 in the Cb of an alcohol-induced cerebellar degeneration mouse model, suggesting that the ZNF212-PLD3 relationship is important for Purkinje cell survival.


Subject(s)
Ataxia/pathology , DNA-Binding Proteins/metabolism , Gait Disorders, Neurologic/pathology , Nerve Tissue Proteins/physiology , Phospholipase D/antagonists & inhibitors , Purkinje Cells/pathology , Animals , Ataxia/etiology , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/genetics , Gait Disorders, Neurologic/etiology , Humans , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Purkinje Cells/metabolism
4.
Dis Model Mech ; 13(7)2020 07 23.
Article in English | MEDLINE | ID: mdl-32540914

ABSTRACT

Peptide therapeutics, unlike small-molecule drugs, display crucial advantages of target specificity and the ability to block large interacting interfaces, such as those of transcription factors. The transcription co-factor of the Hippo pathway, YAP/Yorkie (Yki), has been implicated in many cancers, and is dependent on its interaction with the DNA-binding TEAD/Sd proteins via a large Ω-loop. In addition, the mammalian vestigial-like (VGLL) proteins, specifically their TONDU domain, competitively inhibit YAP-TEAD interaction, resulting in arrest of tumor growth. Here, we show that overexpression of the TONDU peptide or its oral uptake leads to suppression of Yki-driven intestinal stem cell tumors in the adult Drosophila midgut. In addition, comparative proteomic analyses of peptide-treated and untreated tumors, together with chromatin immunoprecipitation analysis, reveal that integrin pathway members are part of the Yki-oncogenic network. Collectively, our findings establish Drosophila as a reliable in vivo platform to screen for cancer oral therapeutic peptides and reveal a tumor suppressive role for integrins in Yki-driven tumors.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Antineoplastic Agents/administration & dosage , DNA-Binding Proteins/administration & dosage , Drosophila melanogaster/drug effects , Drug Development , Intestinal Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Peptide Fragments/administration & dosage , Transcription Factors/administration & dosage , Administration, Oral , Animals , Animals, Genetically Modified , Antineoplastic Agents/metabolism , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic , Humans , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , PC-3 Cells , Peptide Fragments/genetics , Peptide Fragments/metabolism , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
5.
Allergol Immunopathol (Madr) ; 48(6): 654-665, 2020.
Article in English | MEDLINE | ID: mdl-32446781

ABSTRACT

BACKGROUND: The mite alimentary canal contains plenty of microbiota. It is accepted that some of the microbial products function as adjuvants to speed up immune responses. OBJECTIVES: We identified five bacterial proteins from dust mite, and Enterobacterial fimbriae H (FimH) was one of them. This study aims to test a hypothesis that the FimH protein enforces immunotherapy in asthmatic mice. METHODS: Asthmatic mice were treated by allergen specific immunotherapy (ASIT) with rDer f1/f2 or rDer f1/f2 plus FimH. Changes in inflammatory cell infiltration, airway hyperreactivity, frequency of Tregs, splenic CD4+IFN-γ+ cells, and serum levels of TGF-ß, IL-10, IL-13 and IL-17A of asthmatic mice were checked. RESULTS: ASIT with rDer f1/f2 plus FimH reduced inflammatory cell infiltration, airway hyperreactivity (AHR), and levels of IgE and IgG1 compared to ASIT with rDer f1/f2 alone, but the levels of IgG2a increased. Asthmatic mice that underwent ASIT with rDer f1/f2 plus FimH showed increased frequency of Tregs, splenic CD4+IFN-γ+ cells, serum levels of TGF-ß and IL-10; and deceased splenic CD4+IL-4+ cells, and serum levels of IL-13 and IL-17A. In vitro study showed FimH triggered IL-10 expression in a concentration dependent manner and facilitated the differentiation of Tregs. CONCLUSION: Used as an adjuvant, FimH enforces the effect of ASIT in asthmatic mice via augmenting Tregs.


Subject(s)
Asthma/therapy , Bacterial Proteins/administration & dosage , DNA-Binding Proteins/administration & dosage , Desensitization, Immunologic/methods , Enterobacteriaceae/immunology , Pyroglyphidae/microbiology , Adjuvants, Immunologic/administration & dosage , Allergens/administration & dosage , Animals , Antigens, Bacterial/administration & dosage , Antigens, Dermatophagoides/administration & dosage , Antigens, Dermatophagoides/immunology , Asthma/immunology , Cells, Cultured , Dendritic Cells , Disease Models, Animal , Female , Humans , Immunogenicity, Vaccine , Mice , Microbiota/immunology , Primary Cell Culture , Pyroglyphidae/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
6.
Article in English | MEDLINE | ID: mdl-31299884

ABSTRACT

Altered levels of histone acetylation are associated with changes in chromosomal gene expression. Thus, the specific acetylation of histones bound to plasmid DNA might increase transgene expression. Previously, the expression of the histone acetyltransferase domain of CREB-binding protein fused to the sequence-dependent DNA binding domain of GAL4 (GAL4-HAT) successfully improved reporter gene expression in cultured cells [J. Biosci. Bioengng. 123, 277-280 (2017)]. In this study, the same approach was applied for transgene expression in mice. The activator and reporter plasmid DNAs bearing the genes for GAL4-HAT and Gaussia princeps luciferase, respectively, were co-administered into the mouse liver by hydrodynamics-based tail vein injection, and the Gaussia luciferase activity in serum was measured for two weeks. Unexpectedly, the co-injection of the GAL4-HAT and luciferase plasmid DNAs seemed to decrease, rather than increase, luciferase expression. Moreover, the co-injection apparently reduced the amount of luciferase DNA in the liver. These results indicated that this system is ineffective in vivo and suggested the exclusion of hepatic cells expressing GAL4-HAT.


Subject(s)
Histone Acetyltransferases/genetics , Plasmids , Transgenes , Acetylation , Animals , CREB-Binding Protein/genetics , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/genetics , Female , Genes, Reporter , Histone Acetyltransferases/administration & dosage , Luciferases , Mice , Mice, Inbred BALB C , Plasmids/chemistry , Plasmids/genetics , Saccharomyces cerevisiae Proteins/administration & dosage , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/administration & dosage , Transcription Factors/genetics
7.
Neurotherapeutics ; 16(4): 1149-1166, 2019 10.
Article in English | MEDLINE | ID: mdl-31342410

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a debilitating neurodegenerative disorder in which the neuromuscular junction progressively degenerates, leading to movement difficulties, paralysis, and eventually death. ALS is currently being treated by only two FDA-approved drugs with modest efficacy in slowing disease progression. Often, the translation of preclinical findings to bedside terminates prematurely as the evaluation of potential therapeutic compounds focuses on a single study or a single animal model. To circumscribe these issues, we screened 3,765 novel small molecule derivatives of pimozide, a recently identified repurposed neuroleptic for ALS, in Caenorhabditis elegans, confirmed the hits in zebrafish and validated the most active compounds in mouse genetic models. Out of the 27 small molecules identified from the high-throughput screen in worms, 4 were found to recover locomotor defects in C. elegans and genetic zebrafish models of ALS. TRVA242 was identified as the most potent compound as it significantly improved efficiency in rescuing locomotor, motorneuron, and neuromuscular junction synaptic deficits in a C. elegans TDP-43 model and in multiple zebrafish genetic (TDP-43, SOD1, and C9ORF72) models of ALS. The actions of TRVA242 were also conserved in a mammalian model as it also stabilized neuromuscular junction deficits in a mouse SOD1 model of ALS. Compounds such as TRVA242 therefore represent new potential therapeutics for the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , Disease Models, Animal , Neuromuscular Junction/genetics , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/metabolism , Humans , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuromuscular Junction/drug effects , Neuromuscular Junction/metabolism , Organ Culture Techniques , Pimozide/administration & dosage , Pimozide/metabolism , Zebrafish
8.
Chem Biol Drug Des ; 94(3): 1672-1679, 2019 09.
Article in English | MEDLINE | ID: mdl-31108023

ABSTRACT

Thrombotic diseases are major cause of cardiovascular diseases. This study was designed to investigate the effect of tachyplesin I on platelet aggregation and thrombosis. Platelet aggregation was analysed with a whole blood aggregometer. The mice were employed to investigate the effect of tachyplesin I on thrombosis in vivo. Tachyplesin I inhibited thrombin-induced platelet aggregation in a dose-dependent manner. Furthermore, tachyplesin I significantly reduced thrombosis in carrageenan-induced tail thrombosis model by intraperitoneal injection (0.1, 0.2 or 0.4 mg/kg) or intragastric administration (15, 30 or 60 mg/kg). Tachyplesin I also prolonged the bleeding time (BT) and clotting time (CT). The results revealed that tachyplesin I inhibited platelet aggregation and thrombosis by interfering the PI3K/AKT pathway. Tachyplesin I did not show significantly toxicity to mice under 300 mg/kg via intravenous injection. The results show that tachyplesin I inhibits thrombosis and has low toxicity. It is suggested that tachyplesin I has the potential to develop a new anti-thrombotic drug.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , DNA-Binding Proteins/chemistry , Peptides, Cyclic/chemistry , Platelet Aggregation Inhibitors/chemistry , Thrombosis/drug therapy , Animals , Antimicrobial Cationic Peptides/administration & dosage , Bleeding Time , Blood Platelets/metabolism , DNA-Binding Proteins/administration & dosage , Dose-Response Relationship, Drug , Drug Development , Humans , Mice , Models, Animal , Peptides, Cyclic/administration & dosage , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Thrombin/metabolism
9.
J Chemother ; 31(1): 23-29, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30773131

ABSTRACT

This research describes a thermally responsive elastin-like polypeptide (ELP) for the delivery of dnMAML peptides that inhibit the Notch pathway. Exploiting passive targeting and a thermally active tumor-targeting technique available through the use of ELP, the dnMAML peptide was efficiently delivered to tumor tissue. Furthermore, this ELP-dnMAML was modified with the addition of a cell penetrating peptide (SynB1) for improved infiltration of ELP-dnMAML into the tumor cells. In this study, we verified that intravenously delivered SynB1-ELP-dnMAML was cleared from circulation under physiological conditions (37 °C) but accumulated at tumors grown in mice at sites to which an externally induced, local heat (40-41 °C) was applied, thereby resulting in greatly reduced tumor growth in animals. Additionally, in combination with Taxol, SynB1-ELP-dnMAML showed more potent tumor growth retardation.


Subject(s)
DNA-Binding Proteins/administration & dosage , Drug Delivery Systems , Mammary Neoplasms, Experimental/pathology , Paclitaxel/administration & dosage , Peptides/administration & dosage , Receptors, Notch/antagonists & inhibitors , Transcription Factors/administration & dosage , Animals , Antineoplastic Agents, Phytogenic , Cell Line, Tumor , Cell-Penetrating Peptides , Female , Humans , Hyperthermia, Induced , Mice , Mice, Nude , Xenograft Model Antitumor Assays
10.
Mol Cancer Ther ; 18(2): 323-334, 2019 02.
Article in English | MEDLINE | ID: mdl-30401695

ABSTRACT

Renal carcinoma presents a rapid progression in patients with high metastasis with no effective therapeutic strategy. In this study, we designed a folate-grafted PEI600-CyD (H1) nanoparticle-mediated DNA vaccine containing an adjuvant of absent in melanoma 2 (AIM2) and a tumor-specific antigen of carbonic anhydrase IX (CAIX) for renal carcinoma therapy. Mice bearing subcutaneous human CAIX (hCAIX)-Renca tumor were intramuscularly immunized with H1-pAIM2/pCAIX, H1-pCAIX, H1-pAIM2, or Mock vaccine, respectively. The tumor growth of hCAIX-Renca was significantly inhibited in H1-pAIM2/pCAIX vaccine group compared with the control group. The vaccine activated CAIX-specific CD8+ T-cell proliferation and CTL responses, and enhanced the induction of multi-functional CD8+ T cells (expressing TNF-α, IL-2, and IFN-γ). CD8+ T-cell depletion resulted in the loss of anti-tumor activity of H1-pAIM2/pCAIX vaccine, suggesting that the efficacy of the vaccine was dependent on CD8+ T-cell responses. Lung metastasis of renal carcinoma was also suppressed by H1-pAIM2/pCAIX vaccine treatment accompanied with the increased percentages of CAIX-specific multi-functional CD8+ T cells in the spleen, tumor, and bronchoalveolar lavage as compared with H1-pCAIX vaccine. Similarly, the vaccine enhanced CAIX-specific CD8+ T-cell proliferation and CTL responses. Therefore, these results indicated that H1-pAIM2/pCAIX vaccine exhibits the therapeutic efficacy of anti-renal carcinoma by enhancing tumor-specific multi-functional CD8+ T-cell responses. This vaccine strategy could be a potential and promising approach for the therapy of primary solid or metastasis tumors.


Subject(s)
Antigens, Neoplasm/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Carbonic Anhydrase IX/administration & dosage , DNA-Binding Proteins/administration & dosage , Kidney Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Vaccines, DNA/administration & dosage , Animals , Antigens, Neoplasm/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/administration & dosage , Cancer Vaccines/pharmacology , Carbonic Anhydrase IX/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/metabolism , DNA-Binding Proteins/pharmacology , Humans , Injections, Intramuscular , Kidney Neoplasms/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mice , Nanoparticles , Vaccines, DNA/pharmacology , Xenograft Model Antitumor Assays
11.
Nutrients ; 10(12)2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30513901

ABSTRACT

Nesfatin-1 is a well-established anorexigenic peptide. Recent studies indicated an association between nesfatin-1 and anxiety/depression-like behavior. However, it is unclear whether this effect is retained in obesity. The aim was to investigate the effect of nesfatin-130-59-the active core of nesfatin-1-on anxiety and depression-like behavior in normal weight (NW) and diet-induced (DIO) obese rats. Male rats were intracerebroventricularly (ICV) cannulated and received nesfatin-130-59 (0.1, 0.3, or 0.9 nmol/rat) or vehicle 30 min before testing. Nesfatin-130-59 at a dose of 0.3 nmol reduced sucrose consumption in the sucrose preference test in NW rats compared to vehicle (⁻33%, p < 0.05), indicating depression-like/anhedonic behavior. This dose was used for all following experiments. Nesfatin-130-59 also reduced cookie intake during the novelty-induced hypophagia test (-62%, p < 0.05). Moreover, nesfatin-130-59 reduced the number of entries into the center zone in the open field test (-45%, p < 0.01) and the visits of open arms in the elevated zero maze test (-39%, p < 0.01) in NW rats indicating anxiety. Interestingly, DIO rats showed no behavioral alterations after the injection of nesfatin-130-59 (p > 0.05). These results indicate an implication of nesfatin-130-59 in the mediation of anxiety and depression-like behavior/anhedonia under normal weight conditions, while in DIO rats, a desensitization might occur.


Subject(s)
Anhedonia/drug effects , Anxiety/chemically induced , Calcium-Binding Proteins/adverse effects , Calcium-Binding Proteins/chemistry , DNA-Binding Proteins/adverse effects , DNA-Binding Proteins/chemistry , Depression/chemically induced , Nerve Tissue Proteins/adverse effects , Nerve Tissue Proteins/chemistry , Peptide Fragments/adverse effects , Peptide Fragments/chemistry , Animals , Calcium-Binding Proteins/administration & dosage , DNA-Binding Proteins/administration & dosage , Dose-Response Relationship, Drug , Feeding Behavior , Injections, Intraventricular , Male , Nerve Tissue Proteins/administration & dosage , Nucleobindins , Obesity , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley
12.
Peptides ; 109: 14-22, 2018 11.
Article in English | MEDLINE | ID: mdl-30261207

ABSTRACT

Nesfatin-1 is an 82-amino acid protein derived from nucleobindin 2 (NUCB2), which could inhibit food intake in fish and mammals. However, the neuroendocrine mechanism of nesfatin-1 in animal appetite regulation is unclear. To explore the feeding mechanism of nesfatin-1 in Siberian sturgeon (Acipenser baerii), intraperitoneal injections of nesfatin-1 and sulfated cholecystokinin octapeptide (CCK8), Lorglumide (CCK1R selective antagonist), or LY 225,910 (CCK2R selective antagonist) were performed. Co-injection of nesfatin-1 and CCK8 synergistically significantly decreased the food intake in 1 h. Lorglumide reversed the anorectic effect of nesfatin-1, but LY 225,910 had no effect. Moreover, Lorglumide could also reverse the expressions of appetite factors including nucb2, cck, unc3, cart, apelin, pyy, and npy induced by nesfatin-1 in the brain, stomach, and liver, while LY 225,910 partially reversed these changes. These results indicate that nesfatin-1 inhibits the appetite of Siberian sturgeon mainly through the CCK-CCK1R signaling pathway.


Subject(s)
Appetite/drug effects , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Eating/drug effects , Fishes/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Animals , Calcium-Binding Proteins/administration & dosage , Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/pharmacology , Fishes/physiology , Injections, Intraperitoneal , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/pharmacology , Nucleobindins
13.
IUBMB Life ; 70(10): 1002-1011, 2018 10.
Article in English | MEDLINE | ID: mdl-30171788

ABSTRACT

Therapeutic human papillomaviruse (HPV) vaccines have the potential to inhibit the tumor growth by targeting HPV E6 and E7 oncoproteins. Among different vaccine strategies, DNA and protein-based approaches are the most effective candidates for stimulation of the immune responses against HPV infections. Our study was designed to assess the efficacy of small heat shock proteins B1 (Hsp27) and B6 (Hsp20) as an adjuvant accompanied by HPV16 E7 and hPP10-E7 antigens in tumor mouse model. A major key for successful DNA and protein transfer into cells is the development of delivery systems with high efficiency and low cytotoxicity. Herein, we used hPP10 and MPG cell penetrating peptides (CPPs) for protein and DNA delivery in vivo, respectively. Our data indicated that the combination of Hsp27 with the recombinant hPP10-E7 protein in homologous protein/protein (hPP10-E7 + Hsp27) and heterologous DNA/protein (pcDNA-E7 + MPG/ hPP10-E7 + Hsp27) significantly enhanced the E7-specific T cell responses. Indeed, these regimens induced high levels of IgG2a, IFN-γ and IL-2 directed toward Th1 responses and also Granzyme B secretion as compared to other immunization strategies, and also displayed complete protection more than 60 days after treatment. These data suggest that the use of Hsp27 as an adjuvant and MPG and hPP10 as a gene and protein carrier would represent promising applications for improvement of HPV therapeutic vaccines. © 2018 IUBMB Life, 70(10):1002-1011, 2018.


Subject(s)
HSP20 Heat-Shock Proteins/genetics , Heat-Shock Proteins/administration & dosage , Neoplasm Proteins/administration & dosage , Papillomavirus Vaccines/administration & dosage , Uterine Cervical Neoplasms/genetics , Adjuvants, Immunologic/administration & dosage , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , DNA-Binding Proteins/administration & dosage , Female , Granzymes/administration & dosage , Heat-Shock Proteins/genetics , Humans , Molecular Chaperones , Neoplasm Proteins/genetics , Papillomavirus E7 Proteins/administration & dosage , Papillomavirus Vaccines/genetics , Papillomavirus Vaccines/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology
14.
J Cell Mol Med ; 22(11): 5670-5681, 2018 11.
Article in English | MEDLINE | ID: mdl-30160343

ABSTRACT

Renal cell carcinoma (RCC) is a high metastasis tumour with less effective treatment available currently. Absent in melanoma 2 (AIM2) as a tumour suppressor might be used as a potential therapeutic target for RCC treatment. Here, we found that AIM2 expression was significantly decreased in RCC patient specimens and renal carcinoma cell lines (786-O and OSRC-2). To establish a safe and effective AIM2 gene delivery system, we formed the nanoparticles consisting of a folate grafted PEI600-CyD (H1) nanoparticle-mediated AIM2 gene (H1/pAIM2) as an effective delivery agent. Delivery of H1/pAIM2 in renal carcinoma cells could remarkably increase the expression of AIM2, and subsequently decrease cell proliferation, migration, and invasion as well as enhance cell apoptosis. In order to evaluate the therapeutic efficacy of AIM2 in vivo, H1/pAIM2 nanoparticles were injected intratumorally into 786-O-xenograft mice. Administration of H1/pAIM2 nanoparticles could inhibit the tumour growth as evidenced by reduced tumour volume and weight. Furthermore, Blockade of inflammasome activation triggered by H1/pAIM2 nanoparticles using inflammasome inhibitor YVAD-CMK abrogated the anti-tumoral activities of H1/AIM2. These results indicated the therapeutic effect of H1/pAIM2 nanoparticles was mainly attributable to its capability to enhance the inflammasome activation. H1/AIM2 nanoparticles might act as an efficient therapeutic approach for RCC treatment.


Subject(s)
Carcinoma, Renal Cell/therapy , DNA-Binding Proteins/genetics , Gene Transfer Techniques , Nanoparticles/administration & dosage , Animals , Apoptosis/drug effects , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Inflammasomes/genetics , Mice , Nanoparticles/chemistry , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
15.
J Mol Neurosci ; 65(3): 377-384, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29956089

ABSTRACT

Nesfatin-1, a recently discovered peptide, is involved in important functions such as food intake regulation and energy homeostasis. Previous studies have demonstrated that it has protective effects following myocardial injury and also protects dopaminergic cells against neurotoxicity with the anti-inflammatory and anti-apoptotic mechanisms. In this study, we aimed to assay the neuroprotective effects of Nesfatin-1 after brain ischemia/reperfusion. Twenty-eight male Wistar rats were randomly selected and allocated in the form of four groups (sham, Nesfatin-1, ischemia, ischemia+Nesfatin-1). Ischemia was created by obstruction couple common carotid arteries in 20-min period. Saline as a vehicle and Nesfatin-1 (20 µg/kg, intraperitoneally) were injected at the time of reperfusion. Spatial memory performances were evaluated by the Morris water maze. The level of protein expression was determined by immunohistochemical and immunofluorescence staining. Nesfatin-1 significantly reduced caspase-3 (P < 0.01) and microglial activation (P < 0.01) and improved spatial memory impairments (P < 0.05) induced by brain ischemia. Nesfatin-1 has significant neuroprotective effects and can be introduced as a therapeutic agent against cerebral ischemia-induced injuries.


Subject(s)
Brain Ischemia/drug therapy , Calcium-Binding Proteins/therapeutic use , DNA-Binding Proteins/therapeutic use , Memory , Microglia/drug effects , Nerve Tissue Proteins/therapeutic use , Neuroprotective Agents/therapeutic use , Reperfusion Injury/drug therapy , Animals , Calcium-Binding Proteins/administration & dosage , Calcium-Binding Proteins/pharmacology , Caspase 3/metabolism , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/pharmacology , Male , Microglia/metabolism , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/pharmacology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Nucleobindins , Rats , Rats, Wistar
16.
Bull Exp Biol Med ; 165(2): 220-224, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29922995

ABSTRACT

We studied combined effect of ß-hairpin antimicrobial peptide tachyplesin I and cytotoxic agent cisplatin on tumor and normal human cell lines. MTT assay and flow cytometry showed that tachyplesin I selectively sensitized cancer cells to cisplatin in specified concentration ratios. In vitro experiments demonstrated that combined use of tachyplesin I and cisplatin allows decreasing the effective dose of the cytostatic thus reducing nonspecific toxicity.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/pharmacology , DNA-Binding Proteins/pharmacology , Neoplasms/pathology , Peptides, Cyclic/pharmacology , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/administration & dosage , Cell Proliferation/drug effects , Cells, Cultured , Cisplatin/administration & dosage , DNA-Binding Proteins/administration & dosage , Dose-Response Relationship, Drug , Drug Interactions , Drug Screening Assays, Antitumor , HEK293 Cells , HL-60 Cells , Humans , Peptides, Cyclic/administration & dosage
17.
Neuropeptides ; 70: 9-15, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29751997

ABSTRACT

Nesfatin-1, a peptide whose receptor is yet to be identified, has been shown to be involved in the modulation of feeding, stress, and metabolic responses. Recently, increasing evidence has supported a modulatory role of nesfatin-1 in cardiovascular activity. We have previously reported that nesfatin-1 causes an increase in blood pressure in normotensive and hypotensive rats by increasing plasma catecholamine, vasopressin, and renin levels. Recent reports suggest that nesfatin-1 may activate the central cholinergic system. However, there is no evidence showing an interaction between central nesfatin-1 and the cholinergic system. Therefore, this study aimed to determine whether the central cholinergic system may have a functional role in the nesfatin-1-induced cardiovascular effect observed in normotensive rats. Intracerebroventricular injection of nesfatin-1 caused short-term increases in mean arterial pressure and heart rate responses including bradycardic/tachycardic phases in normotensive animals. Central injection of nesfatin-1 increased the acetylcholine and choline levels in the posterior hypothalamus, as shown in microdialysis studies. Central pretreatment with the cholinergic muscarinic receptor antagonist atropine and/or nicotinic receptor antagonist mecamylamine blocked nesfatin-1-induced cardiovascular effects. In conclusion, the results show that centrally administered nesfatin-1 produces a pressor effect on blood pressure and heart rate responses including bradycardic/tachycardic phases in normotensive rats. Moreover, according to our findings, the central cholinergic system can modulate nesfatin-1-evoked cardiovascular activity.


Subject(s)
Blood Pressure/drug effects , Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Hypotension/etiology , Nerve Tissue Proteins/pharmacology , Vasoconstrictor Agents/pharmacology , Acetylcholine/metabolism , Animals , Brain/drug effects , Calcium-Binding Proteins/administration & dosage , Catecholamines/metabolism , Cholinergic Agents/pharmacology , DNA-Binding Proteins/administration & dosage , Heart Rate/drug effects , Male , Mecamylamine/blood , Nerve Tissue Proteins/administration & dosage , Nucleobindins , Rats, Sprague-Dawley , Vasopressins/blood
18.
Nanomedicine (Lond) ; 13(4): 353-372, 2018 02.
Article in English | MEDLINE | ID: mdl-29338617

ABSTRACT

AIM: To investigate anticancer activity of the DNA binding domain of SMAR1 (His 5) in vitro and in vivo. MATERIALS & METHODS: His 5 was conjugated to hydrothermally synthesized carbon nanospheres (CNs). Anticancer activity of CNs-His 5 was evaluated in vitro and in vivo. RESULTS: CNs- His 5 significantly reduced cyclin D1 levels in MDA-MB-231 cells. Tumor bearing Balb/c mice injected with CNs-His 5 showed approximately 62% tumor regression and significantly reduced 18FDG uptake. Caspases assay and IHC staining confirmed tumor growth inhibition, which could be attributed to apoptotic, antiproliferative and antiangiogenic activities of His 5. CONCLUSION: DNA binding domain of the SMAR1 protein (His 5) has potent anticancer activity and its CNs mediated delivery could control breast tumor in mice model.


Subject(s)
Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Carbon/chemistry , Cell Cycle Proteins/administration & dosage , DNA-Binding Proteins/administration & dosage , Drug Carriers/chemistry , Nanospheres/chemistry , Nuclear Proteins/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D1/metabolism , DNA-Binding Proteins/metabolism , Drug Liberation , Female , Humans , Mice, Inbred BALB C , Nuclear Proteins/metabolism , Protein Domains , Recombinant Proteins/administration & dosage , Tissue Distribution
19.
Vaccine ; 35(50): 7026-7032, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29102171

ABSTRACT

A highly effective antigen construct for presenting conserved antigen domains is essential to the development of a universal influenza vaccine. We have developed a novel dual-domain nanoparticle fusion protein (DDNFP) which allows independent presentation of two conserved domains. The conserved domains used were from two separate viral surface proteins, M2e of M2 and fusion peptide (FP) or long alpha helix (CD) of HA2. The carrier is a novel nanoparticle protein - the dodecameric DNA binding protein from starved cells (Dps) of bacteria or archaea. Dps was found to be uniquely capable of simultaneous fusion and surface presentation at both N- and C-termini while retaining the ability to form nanoparticles. Thus, DDNFPs with M2e and FP or CD fused at N- and C-termini of Dps from E. coli (EcDps) or other bacteria were first constructed based on the H1 subtype sequences along with corresponding single-domain nanoparticle fusion proteins (SDNFPs). They were expressed at high levels in bacteria and found to form nanoparticles of the expected size (∼9 nm). They were stable against treatment at high temperatures. The DDNFPs (M2e-EcDps-FP and M2e-EcDps-CD) induced strong antibody responses against individual antigen domains and provided full protection against lethal challenge with PR8 virus (H1N1). Importantly, the protection by DDNFPs was synergistically enhanced as compared to SDNFPs. The M2e-EcDps-CD provided an even stronger protection than M2e-EcDps-FP and therefore appeared to be the superior construct. Together, with novel domain combination, enhanced protection and ease of production, this M2e/CD DDNFP could potentially be a highly effective antigen construct for the universal influenza vaccine.


Subject(s)
Drug Carriers/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Nanoparticles/administration & dosage , Recombinant Fusion Proteins/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/genetics , Antigens, Viral/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Matrix Proteins/genetics
20.
Peptides ; 95: 1-9, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28720397

ABSTRACT

The present study was designed to evaluate the cardioprotective effects of nesfatin-1, a novel peptide with anorexigenic properties, in rats with isoproterenol (ISO)-induced myocardial infarction (MI), and to further investigate the role of Akt/GSK-3ß signaling pathway in the protective effect of nesfatin-1. To induce MI, ISO was subcutaneously injected into the rats for two consecutive days at a dosage of 85mg/kg/day. ISO-induced myocardial damage was indicated by elevated levels of cardiac specific troponin-T, enhanced myocardial expression of proinflammatory cytokines (interleukin-1ß, interleukin-6 and tumor necrosis factor-α), and increased number of cells with apoptotic and necrotic appearance in the myocardial tissue. Levels of p-Akt/Akt and p-GSK-3ß/GSK-3ß significantly decreased in heart tissue after ISO-induced MI. However, intraperitoneal administration of nesfatin-1 (10µg/kg/day) elicited a significant cardioprotective activity by lowering the levels of cardiac troponin-T and proinflammatory cytokines, indicating the protective effect of nesfatin-1 against ISO-induced MI. The biochemical findings were further confirmed by histopathological examination, which was demonstrated by reduced number of apoptotic and necrotic cells. Moreover, expressions of p-Akt/Akt and p-GSK-3ß/GSK-3ß in the myocardium of MI group rats were significantly increased by nesfatin-1 administration, suggesting that nesfatin-1, which appears to possess anti-apoptotic and anti-inflammatory properties, may confer protection against ISO-induced MI via an Akt/GSK-3ß-dependent mechanism.


Subject(s)
Calcium-Binding Proteins/administration & dosage , Cardiotonic Agents/administration & dosage , DNA-Binding Proteins/administration & dosage , Heart/drug effects , Myocardial Infarction/drug therapy , Nerve Tissue Proteins/administration & dosage , Animals , Apoptosis/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3 beta/genetics , Heart/physiopathology , Humans , Isoproterenol/toxicity , Myocardial Infarction/chemically induced , Myocardial Infarction/pathology , Nucleobindins , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL