Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
FASEB J ; 35(11): e21950, 2021 11.
Article in English | MEDLINE | ID: mdl-34613638

ABSTRACT

Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders characterized by relapsing intestinal inflammation, but many details of pathogenesis remain to be fully unraveled. Glucocorticoid (GC)-induced leucine zipper (GILZ) is a mediator of the anti-inflammatory effects of GCs, the most powerful drugs for IBD treatment, but they cause several unwanted side effects. The fusion protein TAT-GILZ has been successfully used in some pre-clinical models of inflammatory and autoimmune diseases. To test the efficacy of TAT-GILZ for treating dextran sulfate sodium (DSS)-induced colitis and explore its impact on the gut microbiome, colitis was induced by DSS in C57BL/6J mice and treated with TAT-GILZ or dexamethasone. Various hallmarks of colitis were analyzed, including disease activity index, gut permeability, and expression of pro-inflammatory cytokines and tight junction proteins. TAT-GILZ treatment showed a therapeutic effect when administered after the onset of colitis. Its efficacy was associated with improved gut permeability, as evidenced by zonula occludens-1 and CD74 upregulation in inflamed colonic tissue. TAT-GILZ also ameliorated the changes in the gut microbiota induced by the DSS, thus potentially providing an optimal environment for colonization of the mucosa surface by beneficial bacteria. Overall, our results demonstrated for the first time that TAT-GILZ treatment proved effective after disease onset allowing restoration of gut permeability, a key pathogenic feature of colitis. Additionally, TAT-GILZ restored gut dysbiosis, thereby contributing to healing mechanisms. Interestingly, we found unprecedented effects of exogenous GILZ that did not overlap with those of GCs.


Subject(s)
Colitis/chemically induced , Colitis/drug therapy , Dextran Sulfate/adverse effects , Intestinal Mucosa/metabolism , Permeability/drug effects , Recombinant Fusion Proteins/administration & dosage , Transcription Factors/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Antigens, Differentiation, B-Lymphocyte/metabolism , Colitis/metabolism , Cytokines/metabolism , Dexamethasone/administration & dosage , Disease Models, Animal , Histocompatibility Antigens Class II/metabolism , Intestinal Mucosa/drug effects , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Trans-Activators/genetics , Transcription Factors/genetics , Treatment Outcome , Up-Regulation/drug effects , Zonula Occludens-1 Protein/metabolism
2.
Parasit Vectors ; 14(1): 209, 2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33879250

ABSTRACT

BACKGROUND: Anopheles arabiensis is an opportunistic malaria vector that rests and feeds outdoors, circumventing current indoor vector control methods. Furthermore, this vector will readily feed on both animals and humans. Targeting this vector while feeding on animals can provide an additional intervention for the current vector control activities. Previous results have displayed the efficacy of using Subolesin/Akirin ortholog vaccines for the control of multiple ectoparasite infestations. This made Akirin a potential antigen for vaccine development against An. arabiensis. METHODS: The efficacy of three antigens, namely recombinant Akirin from An. arabiensis, recombinant Akirin from Aedes albopictus, and recombinant Q38 (Akirin/Subolesin chimera) were evaluated as novel interventions for An. arabiensis vector control. Immunisation trials were conducted based on the concept that mosquitoes feeding on vaccinated balb/c mice would ingest antibodies specific to the target antigen. The antibodies would interact with the target antigen in the arthropod vector, subsequently disrupting its function. RESULTS: All three antigens successfully reduced An. arabiensis survival and reproductive capacities, with a vaccine efficacy of 68-73%. CONCLUSIONS: These results were the first to show that hosts vaccinated with recombinant Akirin vaccines could develop a protective response against this outdoor malaria transmission vector, thus providing a step towards the development of a novel intervention for An. arabiensis vector control.


Subject(s)
Anopheles/immunology , Insect Bites and Stings/immunology , Insect Proteins/immunology , Transcription Factors/immunology , Vaccines/immunology , Animals , Anopheles/genetics , Anopheles/physiology , Female , Humans , Insect Bites and Stings/blood , Insect Bites and Stings/parasitology , Insect Proteins/administration & dosage , Insect Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mosquito Control , Reproduction , Transcription Factors/administration & dosage , Transcription Factors/genetics , Vaccines/administration & dosage , Vaccines/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
3.
Life Sci ; 266: 118886, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33310044

ABSTRACT

AIMS: Triple negative breast cancer (TNBC) has drawn more and more attention due to its high mitotic indices, high metastatic rate and poor prognosis. Gene therapy, especially RNA interference (RNAi), has become a promising targeted therapy. However, improvement of transfection efficiency and discovery of target genes are major problems for the delivery of small interfering RNAs (siRNA). MATERIALS AND METHODS: In the present study, we developed GALA- and CREKA-modified PEG-SS-PEI to deliver siRNAs targeting on EGFR and BRD4 for TNBC therapy. The PEG-SS-PEI/siRNA complexes were prepared by electrostatic interaction and characterized by dynamic light scattering (DLS) and transmission electron microscope (TEM). The release characteristic, stability, cellular uptake and intracellular localization of the complexes were also studied. The effect of the complexes on cell viability was measured in MDA-MB-231 and HUVEC cells. The in vitro anti-tumor activities of the complexes were analyzed by Transwell invasion assay and wound healing assay. The gene silencing effect was evaluated by quantitative real time-polymerase chain reaction (qRT-PCR) and western blot. KEY FINDINGS: The results revealed that the GALA- and CREKA-modified PEG-SS-PEI/siRNA complexes showed excellent transfection efficiency with redox-sensitive release profile and good biological compatibility. The complexes protected siRNA from the degradation of RNA enzymes. The complexes significantly inhibited the proliferation, invasion and migration of MDA-MB-231 cells via the synergistic inhibition of EGFR/PI3K/Akt and BRD4/c-Myc pathways. SIGNIFICANCE: Taken together, co-delivery of siEGFR and siBRD4 by GALA-PEG-SS-PEI and CREKA-PEG-SS-PEI may provide a more effective strategy for the treatment of TNBC.


Subject(s)
Cell Cycle Proteins/administration & dosage , Cell-Penetrating Peptides/chemistry , Gene Silencing , Polyethylene Glycols/chemistry , Polyethyleneimine/analogs & derivatives , RNA, Small Interfering/administration & dosage , Transcription Factors/administration & dosage , Triple Negative Breast Neoplasms/therapy , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Proliferation , ErbB Receptors/administration & dosage , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Genetic Therapy , Humans , Polyethyleneimine/chemistry , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured
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.
Mem Inst Oswaldo Cruz ; 115: e190396, 2020.
Article in English | MEDLINE | ID: mdl-32321154

ABSTRACT

BACKGROUND: Nanoparticles (NPs) are viable candidates as carriers of exogenous materials into cells via transfection and can be used in the DNA vaccination strategy against leptospirosis. OBJECTIVES: We evaluated the efficiency of halloysite clay nanotubes (HNTs) and amine-functionalised multi-walled carbon nanotubes (NH2-MWCNTs) in facilitating recombinant LemA antigen (rLemA) expression and protecting Golden Syrian hamsters (Mesocricetus auratus) against Leptospira interrogans lethal infection. METHODS: An indirect immunofluorescent technique was used to investigate the potency of HNTs and NH2-MWCNTs in enhancing the transfection and expression efficiency of the DNA vaccine in Chinese hamster ovary (CHO) cells. Hamsters were immunised with two doses of vaccines HNT-pTARGET/lemA, NH2-MWCNTs-pTARGET/lemA, pTARGET/lemA, and empty pTARGET (control), and the efficacy was determined in terms of humoral immune response and protection against a lethal challenge. FINDINGS: rLemA DNA vaccines carried by NPs were able to transfect CHO cells effectively, inducing IgG immune response in hamsters (p < 0.05), and did not exhibit cytotoxic effects. Furthermore, 83.3% of the hamsters immunised with NH2-MWCNTs-pTARGET/lemA were protected against the lethal challenge (p < 0.01), and 66.7% of hamsters immunised with HNT-pTARGET/lemA survived (p < 0.05). MAIN CONCLUSIONS: NH2-MWCNTs and HNTs can act as antigen carriers for mammalian cells and are suitable for DNA nanovaccine delivery.


Subject(s)
Antigens, Bacterial/administration & dosage , Bacterial Proteins/administration & dosage , Bacterial Vaccines/administration & dosage , Leptospirosis/prevention & control , Transcription Factors/administration & dosage , Vaccines, DNA/administration & dosage , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Cricetinae , Disease Models, Animal , Female , Fluorescent Antibody Technique, Indirect , Leptospira interrogans/immunology , Leptospirosis/immunology , Nanoparticles , Transcription Factors/immunology , Vaccines, DNA/immunology
6.
Life Sci Alliance ; 3(1)2020 01.
Article in English | MEDLINE | ID: mdl-31843959

ABSTRACT

Myocardial recovery from ischemia-reperfusion (IR) is shaped by the interaction of many signaling pathways and tissue repair processes, including the innate immune response. We and others previously showed that sustained expression of the transcriptional co-activator yes-associated protein (YAP) improves survival and myocardial outcome after myocardial infarction. Here, we asked whether transient YAP expression would improve myocardial outcome after IR injury. After IR, we transiently activated YAP in the myocardium with modified mRNA encoding a constitutively active form of YAP (aYAP modRNA). Histological studies 2 d after IR showed that aYAP modRNA reduced cardiomyocyte (CM) necrosis and neutrophil infiltration. 4 wk after IR, aYAP modRNA-treated mice had better heart function as well as reduced scar size and hypertrophic remodeling. In cultured neonatal and adult CMs, YAP attenuated H2O2- or LPS-induced CM necrosis. TLR signaling pathway components important for innate immune responses were suppressed by YAP/TEAD1. In summary, our findings demonstrate that aYAP modRNA treatment reduces CM necrosis, cardiac inflammation, and hypertrophic remodeling after IR stress.


Subject(s)
Adaptor Proteins, Signal Transducing/administration & dosage , Cardiomegaly/drug therapy , Cardiomegaly/etiology , Myocardial Reperfusion Injury/complications , Myocarditis/drug therapy , Myocarditis/etiology , RNA, Messenger/administration & dosage , Transcription Factors/administration & dosage , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Humans , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Myocardium/immunology , Myocytes, Cardiac/metabolism , Neutrophil Infiltration/drug effects , RNA Editing , RNA, Messenger/genetics , Transcription Factors/genetics , YAP-Signaling Proteins
7.
Mem. Inst. Oswaldo Cruz ; 115: e190396, 2020. graf
Article in English | LILACS | ID: biblio-1101277

ABSTRACT

BACKGROUND Nanoparticles (NPs) are viable candidates as carriers of exogenous materials into cells via transfection and can be used in the DNA vaccination strategy against leptospirosis. OBJECTIVES We evaluated the efficiency of halloysite clay nanotubes (HNTs) and amine-functionalised multi-walled carbon nanotubes (NH2-MWCNTs) in facilitating recombinant LemA antigen (rLemA) expression and protecting Golden Syrian hamsters (Mesocricetus auratus) against Leptospira interrogans lethal infection. METHODS An indirect immunofluorescent technique was used to investigate the potency of HNTs and NH2-MWCNTs in enhancing the transfection and expression efficiency of the DNA vaccine in Chinese hamster ovary (CHO) cells. Hamsters were immunised with two doses of vaccines HNT-pTARGET/lemA, NH2-MWCNTs-pTARGET/lemA, pTARGET/lemA, and empty pTARGET (control), and the efficacy was determined in terms of humoral immune response and protection against a lethal challenge. FINDINGS rLemA DNA vaccines carried by NPs were able to transfect CHO cells effectively, inducing IgG immune response in hamsters (p < 0.05), and did not exhibit cytotoxic effects. Furthermore, 83.3% of the hamsters immunised with NH2-MWCNTs-pTARGET/lemA were protected against the lethal challenge (p < 0.01), and 66.7% of hamsters immunised with HNT-pTARGET/lemA survived (p < 0.05). MAIN CONCLUSIONS NH2-MWCNTs and HNTs can act as antigen carriers for mammalian cells and are suitable for DNA nanovaccine delivery.


Subject(s)
Animals , Female , Bacterial Proteins/administration & dosage , Transcription Factors/administration & dosage , Bacterial Vaccines/administration & dosage , Vaccines, DNA/administration & dosage , Leptospirosis/prevention & control , Antigens, Bacterial/administration & dosage , Bacterial Proteins/immunology , Transcription Factors/immunology , Bacterial Vaccines/immunology , Cricetinae , Fluorescent Antibody Technique, Indirect , Vaccines, DNA/immunology , Disease Models, Animal , Nanoparticles , Leptospira interrogans/immunology , Leptospirosis/immunology , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology
8.
Nutrients ; 12(1)2019 Dec 23.
Article in English | MEDLINE | ID: mdl-31878000

ABSTRACT

Bile acids play an active role in fat metabolism and, in high-fat diets, elevated concentrations of fecal bile acids may be related to an increased risk of colorectal cancer. This study investigated concentrations of fecal and serum bile acids in 36 vegans and 36 omnivores. The reduced rank regression was used to identify dietary patterns associated with fecal bile acids. Dietary patterns were derived with secondary and conjugated fecal bile acids as response variables and 53 food groups as predictors. Vegans had higher fiber (p < 0.01) and lower fat (p = 0.0024) intake than omnivores. In serum, primary and glycine-conjugated bile acids were higher in vegans than in omnivores (p ≤ 0.01). All fecal bile acids were significantly lower in vegans compared to omnivores (p < 0.01). Processed meat, fried potatoes, fish, margarine, and coffee contributed most positively, whereas muesli most negatively to a dietary pattern that was directly associated with all fecal bile acids. According to the pattern, fat intake was positively and fiber intake was inversely correlated with bile acids. The findings contribute to the evidence that, in particular, animal products and fat may play a part in higher levels of fecal bile acids.


Subject(s)
Bile Acids and Salts/analysis , Bile Acids and Salts/blood , Diet, Vegan , Diet , Feces/chemistry , Adult , Animals , Coffee , Cross-Sectional Studies , Dietary Fiber/administration & dosage , Fishes , Glycine/blood , Humans , Male , Margarine , Meat , Middle Aged , Transcription Factors/administration & dosage , Vegans
9.
Respir Med ; 158: 42-48, 2019.
Article in English | MEDLINE | ID: mdl-31605920

ABSTRACT

PURPOSE: We investigated the adverse events (AEs) and treatment completion rates of a 3 month course of once-weekly isoniazid and rifapentine (3H1P1) in South Korean health care workers (HCWs) with latent tuberculosis infection (LTBI). METHODS: HCWs who were candidates for LTBI treatment were enrolled from two tertiary referral centers between December 2016 and October 2017. From December 2016 through March 2017, HCWs who agreed were treated with the 3H1P1 regimen (3H1P1 group). Their compliance and AEs were prospectively collected. From April 2017 onward, HCWs who required LTBI treatment received 3 months of isoniazid plus rifampin (3HR group), and their medical records were retrospectively reviewed. RESULTS: During the study period, 406 HCWs were treated, 226 (55.7%) in the 3H1P1 group, and 180 (44.3%) in the 3HR group. The number of subjects with AEs was significantly greater in the 3H1P1 group (75.2% vs 56.7%, P < 0.001), in particular a flu-like syndrome (19.0% vs. 0%, P < 0.001). However, hepatotoxicity occurred less frequently in those receiving 3H1P1 (7.5% vs. 20.0%, P < 0.001). Per protocol definition, anaphylaxis developed in 1.8% of the 3H1P1 group. The overall treatment completion rate was greater in the 3H1P1 group (92.9% vs 86.7%, P = 0.036). CONCLUSIONS: The 3H1P1 regimen had a higher treatment completion rate and lower hepatotoxicity compared with the 3HR regimen. However, it resulted in a higher rate of flu-like syndromes. Additionally, a few subjects had anaphylaxis, although there were no fatalities.


Subject(s)
Antitubercular Agents/administration & dosage , Health Personnel , Isoniazid/administration & dosage , Isoniazid/adverse effects , Latent Tuberculosis/drug therapy , RNA-Binding Proteins/administration & dosage , RNA-Binding Proteins/adverse effects , Transcription Factors/administration & dosage , Transcription Factors/adverse effects , Anaphylaxis/chemically induced , Antitubercular Agents/adverse effects , Female , Humans , Male , Occupational Health , Republic of Korea , Time Factors
10.
PLoS One ; 14(9): e0220684, 2019.
Article in English | MEDLINE | ID: mdl-31479462

ABSTRACT

Bacterial resistance to antibiotics is widely regarded as a major public health concern with last resort MRSA treatments like vancomycin now encountering resistant strains. TFDs (Transcription Factor Decoys) are oligonucleotide copies of the DNA-binding sites for transcription factors. They bind to and sequester the targeted transcription factor, thus inhibiting transcription of many genes. By developing TFDs with sequences aimed at inhibiting transcription factors controlling the expression of highly conserved bacterial cell wall proteins, TFDs present as a potential method for inhibiting microbial growth without encountering typical resistance mechanisms. However, the efficient protection and delivery of the TFDs inside the bacterial cells is a critical step for the success of this technology. Therefore, in our study, specific TFDs against S. aureus were complexed with two different types of nanocarriers: cationic nanostructured lipid carriers (cNLCs) and chitosan-based nanoparticles (CS-NCs). These TFD-carrier nanocomplexes were characterized for size, zeta potential and TFD complexation or loading efficiency in a variety of buffers. In vitro activity of the nanocomplexes was examined alone and in combination with vancomycin, first in methicillin susceptible strains of S. aureus with the lead candidate advancing to tests against MRSA cultures. Results found that both cNLCs and chitosan-based carriers were adept at complexing and protecting TFDs in a range of physiological and microbiological buffers up to 72 hours. From initial testing, chitosan-TFD particles demonstrated no visible improvements in effect when co-administered with vancomycin. However, co-delivery of cNLC-TFD with vancomycin reduced the MIC of vancomycin by over 50% in MSSA and resulted in significant decreases in viability compared with vancomycin alone in MRSA cultures. Furthermore, these TFD-loaded particles demonstrated very low levels of cytotoxicity and haemolysis in vitro. To our knowledge, this is the first attempt at a combined antibiotic/oligonucleotide-TFD approach to combatting MRSA and, as such, highlights a new avenue of MRSA treatment combining traditional small molecules drugs and bacterial gene inhibition.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Lipids , Methicillin-Resistant Staphylococcus aureus/drug effects , Nanostructures , Transcription Factors/administration & dosage , Vancomycin/administration & dosage , Anti-Bacterial Agents/chemistry , Chitosan/chemistry , Drug Carriers , Drug Compounding , Drug Delivery Systems , Drug Stability , Drug Synergism , Hemolysis/drug effects , Humans , Lipids/chemistry , Methicillin-Resistant Staphylococcus aureus/genetics , Microbial Sensitivity Tests , Models, Biological , Nanostructures/chemistry , Staphylococcal Infections/microbiology , Transcription Factors/chemistry
11.
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
12.
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
13.
Methods Mol Biol ; 1867: 239-251, 2018.
Article in English | MEDLINE | ID: mdl-30155828

ABSTRACT

Artificial transcription factors based on zinc finger, TALE, and CRISPR/Cas9 programmable DNA-binding platforms have been widely used to regulate the expression of specific genes in cultured cells, but their delivery into organs such as the brain represents a critical challenge to apply such tools in live animals. In previous work, we developed a zinc-finger-based artificial transcription factor harboring a cell-penetrating peptide (CPP) that could be injected systemically, cross the blood-brain barrier, and alter expression of a specific gene in the brain of an adult mouse. Importantly, our mode of delivery produced widespread distribution throughout the brain. Here we describe methods for the production and purification of the factor, testing CPP activity in cells, and testing CPP activity in mice.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/metabolism , Cell-Penetrating Peptides/administration & dosage , Gene Transfer Techniques , Genetic Engineering/methods , Transcription Factors/administration & dosage , Zinc Fingers , Animals , Cell-Penetrating Peptides/genetics , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Transcription Factors/genetics
14.
Elife ; 72018 05 01.
Article in English | MEDLINE | ID: mdl-29714688

ABSTRACT

Identification of optimal transcription factor expression patterns to direct cellular differentiation along a desired pathway presents significant challenges. We demonstrate massively combinatorial screening of temporally-varying mRNA transcription factors to direct differentiation of neural progenitor cells using a dynamically-reconfigurable magnetically-guided spotting technology for localizing mRNA, enabling experiments on millimetre size spots. In addition, we present a time-interleaved delivery method that dramatically reduces fluctuations in the delivered transcription factor copy numbers per cell. We screened combinatorial and temporal delivery of a pool of midbrain-specific transcription factors to augment the generation of dopaminergic neurons. We show that the combinatorial delivery of LMX1A, FOXA2 and PITX3 is highly effective in generating dopaminergic neurons from midbrain progenitors. We show that LMX1A significantly increases TH-expression levels when delivered to neural progenitor cells either during proliferation or after induction of neural differentiation, while FOXA2 and PITX3 increase expression only when delivered prior to induction, demonstrating temporal dependence of factor addition.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Dopaminergic Neurons/cytology , Embryonic Stem Cells/cytology , Magnetics , Neural Stem Cells/cytology , RNA, Messenger/administration & dosage , Cells, Cultured , Dopaminergic Neurons/metabolism , Drug Delivery Systems , Embryonic Stem Cells/metabolism , Hepatocyte Nuclear Factor 3-beta/administration & dosage , Hepatocyte Nuclear Factor 3-beta/genetics , Homeodomain Proteins/administration & dosage , Homeodomain Proteins/genetics , Humans , LIM-Homeodomain Proteins/administration & dosage , LIM-Homeodomain Proteins/genetics , Neural Stem Cells/metabolism , RNA, Messenger/genetics , Transcription Factors/administration & dosage , Transcription Factors/genetics
15.
PLoS One ; 13(4): e0195558, 2018.
Article in English | MEDLINE | ID: mdl-29617431

ABSTRACT

Delivery of recombinant proteins to therapeutic cells is limited by a lack of efficient methods. This hinders the use of transcription factors or Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) ribonucleoproteins to develop cell therapies. Here, we report a soluble peptide designed for the direct delivery of proteins to mammalian cells including human stem cells, hard-to-modify primary natural killer (NK) cells, and cancer cell models. This peptide is composed of a 6x histidine-rich domain fused to the endosomolytic peptide CM18 and the cell penetrating peptide PTD4. A less than two-minute co-incubation of 6His-CM18-PTD4 peptide with spCas9 and/or asCpf1 CRISPR ribonucleoproteins achieves robust gene editing. The same procedure, co-incubating with the transcription factor HoxB4, achieves transcriptional regulation. The broad applicability and flexibility of this DNA- and chemical-free method across different cell types, particularly hard-to-transfect cells, opens the way for a direct use of proteins for biomedical research and cell therapy manufacturing.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Transcription Factors/administration & dosage , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cricetulus , Cytosol/metabolism , Endocytosis , Escherichia coli , Humans , Mice , Myoblasts/cytology , Myoblasts/metabolism , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism
16.
Methods Mol Biol ; 1767: 227-239, 2018.
Article in English | MEDLINE | ID: mdl-29524138

ABSTRACT

The ability to activate or repress specific genes in the brain could have a tremendous impact for understanding and treating neurological disorders. Artificial transcription factors based on zinc finger, TALE, and CRISPR/Cas9 programmable DNA-binding platforms have been widely used to regulate the expression of specific genes in cultured cells, but their delivery into the brain represents a critical challenge to apply such tools in live animals. In previous work, we developed a purified, zinc finger-based artificial transcription factor that could be injected systemically, cross the blood-brain barrier, and alter expression of a specific gene in the brain of an adult mouse model of Angelman syndrome. Importantly, our mode of delivery produced widespread distribution throughout the brain. Here we describe our most current methods for the production and purification of the factor, dosage optimization, and use of live animal fluorescence imaging to visualize the kinetics of distribution.


Subject(s)
Alleles , Brain/metabolism , Gene Silencing , Transcription Factors/administration & dosage , Transcriptional Activation , Animals , Injections , Mice , Mice, Inbred C57BL , Optical Imaging/methods , Transcription Factors/chemistry , Transcription Factors/pharmacokinetics , Zinc Fingers
17.
J Control Release ; 269: 24-35, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29113792

ABSTRACT

Transcription factors (TFs) are at the center of the broad regulatory network orchestrating gene expression programs that elicit different biological responses. For a long time, TFs have been considered as potent drug targets due to their implications in the pathogenesis of a variety of diseases. At the same time, TFs, located at convergence points of cellular regulatory pathways, are powerful tools providing opportunities both for cell type change and for managing the state of cells. This task formulation requires the TF modulation problem to come to the fore. We review several ways to manage TF activity (small molecules, transfection, nanocarriers, protein-based approaches), analyzing their limitations and the possibilities to overcome them. Delivery of TFs could revolutionize the biomedical field. Whether this forecast comes true will depend on the ability to develop convenient technologies for targeted delivery of TFs.


Subject(s)
Transcription Factors , Animals , Cell Transdifferentiation , DNA , Drug Delivery Systems , Humans , Pluripotent Stem Cells , RNA , Transcription Factors/administration & dosage , Transcription Factors/metabolism
18.
Neuropharmacology ; 126: 1-11, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28823612

ABSTRACT

Therapeutic use of GABAB receptor agonists for conditions like chronic abdominal pain, overactive bladder (OAB) and gastroesophageal reflux disease (GERD) is severely affected by poor blood-brain barrier permeability and potential side effects. ADX71441 is a novel positive allosteric modulator (PAM) of the GABAB receptor that has shown encouraging results in pre-clinical models of anxiety, pain, OAB and alcohol addiction. The present study investigates the analgesic effect of ADX71441 to noxious stimulation of the urinary bladder and colon in rats. In female Sprague-Dawley rats, systemic (i.p), but not intrathecal (i.t), administration of ADX71441 produced a dose-dependent decrease in viscero-motor response (VMR) to graded urinary bladder distension (UBD) and colorectal distension (CRD). Additionally, intra-cerebroventricular (i.c.v.) administration of ADX71441 significantly decreased the VMRs to noxious UBD. In electrophysiology experiments, the drug did not attenuate the responses of UBD-sensitive pelvic nerve afferent (PNA) fibers to UBD. In contrast, ADX71441 significantly decreased the responses of UBD-responsive lumbosacral (LS) spinal neurons in spinal intact rats. However, ADX71441 did not attenuate these LS neurons in cervical (C1-C2) spinal transected rats. During cystometrogram (CMG) recordings, ADX71441 (i.p.) significantly decreased the VMR to slow infusion without affecting the number of voiding contraction. These results indicate that ADX71441 modulate bladder nociception via its effect at the supra-spinal sites without affecting the normal bladder motility and micturition reflex in naïve adult rats.


Subject(s)
Analgesics/administration & dosage , Bacterial Proteins/administration & dosage , Nociception/drug effects , Receptors, GABA-B/physiology , Transcription Factors/administration & dosage , Urinary Bladder/physiopathology , Visceral Pain/prevention & control , Abdominal Oblique Muscles/drug effects , Abdominal Oblique Muscles/physiopathology , Acetamides , Allosteric Regulation , Animals , Colon/physiopathology , Female , Injections, Spinal , Neurons/drug effects , Neurons/physiology , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/physiopathology , Triazines , Urinary Bladder/drug effects
19.
Bioorg Med Chem Lett ; 27(15): 3542-3545, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28596054

ABSTRACT

The natriuretic peptide (NP) system is a critical endocrine, autocrine, and paracrine system and has been investigated for potential use against cardiovascular and metabolic diseases. The clearance of NPs is regulated by the proteolysis of neutral endopeptidase (NEP) and by endocytosis via natriuretic peptide receptor-3 (NPR3). A linear NPR3-selective peptide, [Cha8]-ANP(7-16)-NH2 (1), showed potent binding affinity for NPR3 but poor predicted chemical stability due to its free thiol group. A 12-mer peptide (9) without a thiol group was designed by the hybridization of two NPR3-binding peptides: a linear ANP fragment peptide analog and musclin, a murine member of the bHLH family of transcription factors, possessed high binding affinity and strict selectivity for NPR3. To increase the proteolytic resistance of 9, amino acid substitutions at the cleavage sites led to hydroxyacetyl-[d-Phe5,d-Hyp7,Cha8,d-Ser9,Hyp11,Arg(Me)14]-ANP(5-15)-NHCH3 (23), showing high and selective binding affinity for NPR3 over NPR1 and excellent stability in mouse serum. Compound 23 increased intracellular cGMP concentrations in primary cultured adipocytes, and continuous administration induced substantial plasma cGMP elevation in mice, suggesting its potential to clarify the physiological role of NPR3 and its therapeutic application.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Muscle Proteins/pharmacology , Peptide Fragments/pharmacology , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Transcription Factors/pharmacology , Amino Acid Sequence , Animals , Atrial Natriuretic Factor/administration & dosage , Atrial Natriuretic Factor/blood , Atrial Natriuretic Factor/chemistry , Mice , Mice, Inbred C57BL , Muscle Proteins/administration & dosage , Muscle Proteins/blood , Muscle Proteins/chemistry , Peptide Fragments/administration & dosage , Peptide Fragments/blood , Peptide Fragments/chemistry , Receptors, Atrial Natriuretic Factor/metabolism , Transcription Factors/administration & dosage , Transcription Factors/blood , Transcription Factors/chemistry
20.
Biochem Pharmacol ; 144: 29-34, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28642036

ABSTRACT

Targeting transcription factors represents one possibility to interfere with a known activated regulatory pathway that promotes disease. Double-stranded transcription factor decoy (TFD) oligodeoxynucleotides (ODN) are therapeutic drug candidates, which are able to specifically target and neutralize key transcription factors involved in the pathogenesis of a given disease. These short double-stranded TFD molecules mimic the consensus DNA binding site of a specific transcription factor in the promoter region of its target genes. Therefore, it is possible to exploit this nucleic acid-based drug class for the treatment of diseases caused by aberrant expression of such target genes the products of which are involved in disease initiation and progression. This research update focuses firstly on the mechanism of action of TFD molecules. Long-term effects of such ODNs depend on their stability and the efficiency by which they are delivered to the target tissue and taken up by their target cells. Hence structural modifications like e.g., single-stranded TFD molecules hybridising to itself to form an intramolecular hairpin molecule or circular ODNs assuming a dumbbell configuration, intended to enhance both stability and efficacy, are addressed. Also specific drug delivery methods like ultrasound-targeted microbubble destruction with TFD ODN-coated microbubbles or adeno-associated viral (AAV) vectors for tissue-specific transduction and long-term TFD molecule expression in non-dividing cells will be discussed. Finally, current therapeutic applications of TFD ODN will be summarized.


Subject(s)
Drug Delivery Systems/trends , Oligodeoxyribonucleotides/administration & dosage , Technology, Pharmaceutical/trends , Transcription Factors/administration & dosage , Animals , Binding Sites/physiology , Clinical Trials as Topic/methods , Dermatitis/drug therapy , Dermatitis/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Oligodeoxyribonucleotides/metabolism , Technology, Pharmaceutical/methods , Transcription Factors/metabolism , Vascular Diseases/drug therapy , Vascular Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...