Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 349
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Pharm ; 21(10): 5217-5237, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39185610

ABSTRACT

Nimodipine is the primary clinical drug used to treat cerebral vasospasm following subarachnoid hemorrhage. Currently, tablets have low bioavailability when taken orally, and injections contain ethanol. Therefore, we investigated a new method of nimodipine administration, namely, nasoencephalic administration. Nasal administration of nimodipine was carried out by attaching the cell-penetrating peptide octa-arginine (R8) to liposomes of nimodipine and incorporating it into a temperature-sensitive in situ gel. The prepared liposomes and gels underwent separate evaluations for in vitro characterization. In vitro release exhibited a significant slow-release effect. In vitro toad maxillary cilia model, RPMI 2650 cytotoxicity, and in vivo SD rat pathological histotoxicity experiments showed that all the dosage from the groups had no significant toxicity to toad maxillary cilia, RPMI 2650 cells, and SD rat tissues and organs, and the cilia continued to oscillate up to 694 ± 10.15 min, with the survival rate of the cells being above 85%. A transwell nasal mucosa cell model and an isolated porcine nasal mucosa model were established, and the results showed that the osmolality of the R8-modified nimodipine liposomal gel to nasal mucosal cells and isolated porcine nasal mucosa was 30.41 ± 2.14 and 65.9 ± 7.34 µg/mL, respectively, which was significantly higher than that of the NM-Solution and PEGylated nimodipine liposome gel groups. Animal fluorescence imaging studies revealed that the R8-modified nimodipine liposomal gel displayed increased brain fluorescence intensity compared to the normal liposomal gel. Pharmacokinetic results showed that after transnasal administration, the AUC(0-∞) of the R8-modified nimodipine liposomal gel was 11.662 ± 1.97 µg·mL-1, which was significantly higher than that of the plain nimodipine liposomal gel (5.499 ± 2.89 µg·mL-1). Brain-targeting experiments showed that the brain-targeting efficiencies of the PEGylated nimodipine liposome gel and R8-modified PEGylated nimodipine liposome gels were 20.44 and 33.45, respectively, suggesting that R8/PEG/Lip-NM-TSG significantly increased the brain-targeting of the drug.


Subject(s)
Administration, Intranasal , Gels , Liposomes , Nimodipine , Rats, Sprague-Dawley , Animals , Nimodipine/administration & dosage , Nimodipine/chemistry , Nimodipine/pharmacokinetics , Rats , Liposomes/chemistry , Gels/chemistry , Male , Nasal Mucosa/metabolism , Nasal Mucosa/drug effects , Swine , Arginine/chemistry , Cilia/drug effects , Temperature , Drug Delivery Systems/methods , Humans , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/administration & dosage , Anura , Cell Line
2.
Mol Pharm ; 21(5): 2097-2117, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38440998

ABSTRACT

Currently, one of the most significant and rapidly growing unmet medical challenges is the treatment of neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). This challenge encompasses the imperative development of efficacious therapeutic agents and overcoming the intricacies of the blood-brain barrier for successful drug delivery. Here we focus on the delivery aspect with particular emphasis on cell-penetrating peptides (CPPs), widely used in basic and translational research as they enhance drug delivery to challenging targets such as tissue and cellular compartments and thus increase therapeutic efficacy. The combination of CPPs with nanomaterials such as nanoparticles (NPs) improves the performance, accuracy, and stability of drug delivery and enables higher drug loads. Our review presents and discusses research that utilizes CPPs, either alone or in conjugation with NPs, to mitigate the pathogenic effects of neurodegenerative diseases with particular reference to AD and PD.


Subject(s)
Blood-Brain Barrier , Cell-Penetrating Peptides , Drug Delivery Systems , Nanoparticles , Neurodegenerative Diseases , Parkinson Disease , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/administration & dosage , Humans , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Drug Delivery Systems/methods , Nanoparticles/chemistry , Neurodegenerative Diseases/drug therapy , Animals , Parkinson Disease/drug therapy , Alzheimer Disease/drug therapy
3.
Biol Pharm Bull ; 47(5): 1033-1042, 2024.
Article in English | MEDLINE | ID: mdl-38797668

ABSTRACT

Eye drops, including solutions and suspensions, are essential dosage forms to treat ophthalmic diseases, with poorly water-soluble drugs typically formulated as ophthalmic suspensions. In addition to low bioavailability, suspensions exhibit limited efficacy, safety, and usability due to the presence of drug particles. Improving bioavailability can reduce the drug concentrations and the risk of problems associated with suspended drug particles. However, practical penetration enhancers capable of improving bioavailability remain elusive. Herein, we focused on penetratin (PNT), a cell-penetrating peptide (CPP) that promotes active cellular transport related to macromolecule uptake, such as micropinocytosis. According to the in vitro corneal uptake study using a reconstructed human corneal epithelial tissue model, LabCyte CORNEA-MODEL24, PNT enhanced the uptake of Fluoresbrite® YG carboxylate polystyrene microspheres without covalent binding. In an ex vivo porcine eye model, the addition of 10 µM PNT to rebamipide ophthalmic suspension markedly improved the corneal uptake of rebamipide; however, the addition of 100 µM PNT was ineffective due to potentially increased particle size by aggregation. This article provides basic information on the application of PNT as a penetration enhancer in ophthalmic suspensions, including the in vitro and ex vivo studies mentioned above, as well as the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cytotoxicity assay and storage stability at different pH values.


Subject(s)
Cell-Penetrating Peptides , Cornea , Ophthalmic Solutions , Suspensions , Animals , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/administration & dosage , Ophthalmic Solutions/administration & dosage , Humans , Cornea/metabolism , Cornea/drug effects , Swine , Quinolones/administration & dosage , Quinolones/pharmacokinetics , Quinolones/chemistry , Administration, Ophthalmic , Biological Availability , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Particle Size , Alanine/analogs & derivatives
4.
Am J Hum Genet ; 104(2): 341-347, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30712775

ABSTRACT

Erythropoietic protoporphyria (EPP) is a hereditary disease characterized by a deficiency in ferrochelatase (FECH) activity. FECH activity is responsible for the accumulation of protoporphyrin IX (PPIX). Without etiopathogenic treatment, EPP manifests as severe photosensitivity. 95% of affected individuals present a hypomorphic FECH allele trans to a loss-of-function (LOF) FECH mutation, resulting in a reduction in FECH activity in erythroblasts below a critical threshold. The hypomorphic allele promotes the use of a cryptic acceptor splice site, generating an aberrant FECH mRNA, which is responsible for the reduced level of wild-type FECH mRNA and, ultimately, FECH activity. We have previously identified an antisense oligonucleotide (AON), AON-V1 (V1), that redirects splicing to the physiological acceptor site and reduces the accumulation of PPIX. Here, we developed a specific strategy that uses transferrin receptor 1 (TRF1) as a Trojan horse to deliver V1 to erythroid progenitors. We designed a bifunctional peptide (P1-9R) including a TFR1-targeting peptide coupled to a nine-arginine cell-penetrating peptide (CPP) that facilitates the release of the AON from TFR1 in endosomal vesicles. We demonstrated that the P1-9R/V1 nanocomplex promotes the efficient and prolonged redirection of splicing towards the physiological splice site and subsequent normalization of WT FECH mRNA and protein levels. Finally, the P1-9R/V1 nanocomplex increases WT FECH mRNA production and significantly decreases PPIX accumulation in primary cultures of differentiating erythroid progenitors from an overt EPP-affected individual. P1-9R is a method designed to target erythroid progenitors and represents a potentially powerful tool for the in vivo delivery of therapeutic DNA in many erythroid disorders.


Subject(s)
Antigens, CD/metabolism , Cell-Penetrating Peptides/metabolism , Erythroid Precursor Cells/metabolism , Genetic Therapy/methods , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/therapy , Receptors, Transferrin/metabolism , Antigens, CD/administration & dosage , Antigens, CD34/metabolism , Cell Line , Cell-Penetrating Peptides/administration & dosage , Erythroblasts/cytology , Erythroblasts/metabolism , Ferrochelatase/genetics , Ferrochelatase/metabolism , Humans , Ligands , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Protoporphyrins/metabolism , RNA, Messenger , Receptors, Transferrin/administration & dosage
5.
Mol Ther ; 29(5): 1744-1757, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33545360

ABSTRACT

Cardiovascular disease is the leading cause of death and disability worldwide. Effective delivery of cell-selective therapies that target atherosclerotic plaques and neointimal growth while sparing the endothelium remains the Achilles heel of percutaneous interventions. The current study utilizes synthetic microRNA switch therapy that self-assembles to form a compacted, nuclease-resistant nanoparticle <200 nM in size when mixed with cationic amphipathic cell-penetrating peptide (p5RHH). These nanoparticles possess intrinsic endosomolytic activity that requires endosomal acidification. When administered in a femoral artery wire injury mouse model in vivo, the mRNA-p5RHH nanoparticles deliver their payload specifically to the regions of endothelial denudation and not to the lungs, liver, kidney, or spleen. Moreover, repeated administration of nanoparticles containing a microRNA switch, consisting of synthetically modified mRNA encoding for the cyclin-dependent kinase inhibitor p27Kip1 that contains one complementary target sequence of the endothelial cell-specific miR-126 at its 5' UTR, drastically reduced neointima formation after wire injury and allowed for vessel reendothelialization. This cell-selective nanotherapy is a valuable tool that has the potential to advance the fight against neointimal hyperplasia and atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Cell-Penetrating Peptides/administration & dosage , Cyclin-Dependent Kinase Inhibitor p27/antagonists & inhibitors , Femoral Artery/injuries , MicroRNAs/administration & dosage , Animals , Atherosclerosis/etiology , Cell-Penetrating Peptides/pharmacology , Coronary Restenosis , Disease Models, Animal , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Nanoparticles , Particle Size , Synthetic Biology
6.
Int J Cancer ; 149(6): 1313-1321, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34019700

ABSTRACT

CIGB-552 is a synthetic peptide that interacts with COMMD1 and upregulates its protein levels. The objectives of this phase I study were safety, pharmacokinetic profile, evaluation of the lymphocytes CD4+ and CD8+ and preliminary activity in patients with advanced tumors. A 3 + 3 dose-escalation design with seven dose levels was implemented. Patients were included until a grade 3 related adverse event occurred and the maximum tolerated dose was reached. The patients received subcutaneous administration of CIGB-552 three times per week for 2 weeks. Single-dose plasma pharmacokinetics was characterized at two dose levels, and tumor responses were classified by RECIST 1.1. Twenty-four patients received CIGB-552. Dose-limiting toxicity was associated with a transient grade 3 pruritic maculopapular rash at a dose of 7.0 mg. The maximum tolerated dose was defined as 4.7 mg. Ten patients were assessable for immunological status. Seven patients had significant changes in the ratio CD4/CD8 in response to CIGB-552 treatment; three patients did not modify the immunological status. Stable disease was observed in five patients, including two metastatic soft sarcomas. We conclude that CIGB-552 at dose 4.7 mg was well tolerated with no significant adverse events and appeared to provide some clinical benefits.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell-Penetrating Peptides/administration & dosage , NF-kappa B/drug effects , Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/metabolism , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms/metabolism , Neoplasms/pathology , Research Design , Treatment Outcome
7.
Mol Pharm ; 18(3): 796-806, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33464088

ABSTRACT

The small interference RNA (siRNA)-assisted RNA interference approach in stem cells for differentiating into cell-specific lineages is gaining importance for its therapeutic potential. An effective gene delivery platform is crucial to achieve this goal. In this context, self-fluorescent, cell-penetrating peptide (CPP)-functionalized hydroxyapatite nanoparticles (R8HNPs) were synthesized by a modified sol gel technique. R8HNPs were crystalline, displayed characteristic bands, and exhibited broad emission spectra from 350 to 750 nm corresponding to green and red fluorescence. The biocompatible R8HNPs displayed robust binding with siRNA and excellent uptake in R1 ESCs. This was attributed to functionalization with CPP. Moreover, the R8HNP-complexed siRNA exhibited excellent serum and room temperature stability. The NPs protected the siRNA from sonication, pH, and temperature-induced stress and efficiently delivered siRNA to trigger 80% silencing of a pluripotency marker gene, Oct4, in R1 ESCs at 48 h. The transient downregulation was also observed at the protein level. Our findings demonstrate R8HNPs as a promising delivery agent for siRNA therapeutics with the potential for lineage-specific differentiation and future applications in regenerative medicine.


Subject(s)
Durapatite/chemistry , Mouse Embryonic Stem Cells/drug effects , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Down-Regulation/drug effects , Gene Transfer Techniques , Mice , Nanoparticles/chemistry , RNA Interference/drug effects , RNA, Small Interfering/chemistry
8.
Anticancer Drugs ; 32(2): 178-188, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32826414

ABSTRACT

Ultrasound-mediated nanobubble destruction (UMND), which can utilize the physical energy of ultrasound irradiation to improve the transfer efficiency to target cells is becoming one of the most promising carriers for gene delivery. The purpose of this study was to establish cell-penetrating peptide (CPP)-loaded nanobubbles (CNBs) connected with long intergenic nonprotein coding RNA 00511-small interfering RNA (LINC00511-siRNA) and evaluate its feasibility for improving the chemosensitivity of triple-negative breast cancer in vitro. First, fluorescence imaging confirmed the loading of siLINC00511 on CNBs, and the CNBs-siLINC00511 were characterized by the Zetasizer Nano ZS90 analyzer and transmission electron microscopy. Next, cell counting kit 8 assay was used to detect the inhibitory activity of cisplatin on the proliferation of MDA-MB-231 cells, and the 50% inhibition concentration value before and after transfer was calculated. Finally, the silencing effect of siLINC00511 was evaluated in vitro using an apoptosis assay, transwell assay, real time-PCR and western blotting. UMND combined with CNBs could effectively transfer the siRNA to MDA-MB-231 cells, thus evidently reducing the expression of LINC00511. Furthermore, inhibitory activity of cisplatin on MDA-MB-231 cells was enhanced after downregulation of LINC00511 expression. Downregulation of LINC00511 alters expression of cell cycle-related (CDK 6) and apoptosis-related (Bcl-2 and Bax) proteins in MDA-MB-231 cells. These results suggested that siRNA-CNBs may be an ideal vector for the treatment of tumors, with high efficiency RNA interference under the combined action of UMND. It may provide a new therapeutic method for triple negative breast cancer.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Cisplatin/pharmacology , RNA, Long Noncoding/pharmacology , RNA, Small Interfering/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Ultrasonic Waves , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , Cisplatin/administration & dosage , Cyclin-Dependent Kinase 6/metabolism , Down-Regulation , Female , Genetic Vectors , Humans , Inhibitory Concentration 50 , Nanoparticles/chemistry , Polymers/chemistry , RNA, Long Noncoding/administration & dosage , RNA, Small Interfering/administration & dosage , Triple Negative Breast Neoplasms/pathology
9.
Arterioscler Thromb Vasc Biol ; 40(12): 2990-3003, 2020 12.
Article in English | MEDLINE | ID: mdl-33028101

ABSTRACT

OBJECTIVE: Arterial thrombosis leading to ischemic injury worsens the prognosis of many patients with cardiovascular disease. PZ-128 is a first-in-class pepducin that reversibly inhibits PAR1 (protease-activated receptor 1) on platelets and other vascular cells by targeting the intracellular surface of the receptor. The TRIP-PCI (Thrombin Receptor Inhibitory Pepducin in Percutaneous Coronary Intervention) trial was conducted to assess the safety and efficacy of PZ-128 in patients undergoing cardiac catheterization with intent to perform percutaneous coronary intervention. Approach and Results: In this randomized, double-blind, placebo-controlled, phase 2 trial, 100 patients were randomly assigned (2:1) to receive PZ-128 (0.3 or 0.5 mg/kg), or placebo in a 2-hour infusion initiated just before the start of cardiac catheterization, on top of standard oral antiplatelet therapy. Rates of the primary end point of bleeding were not different between the combined PZ-128 doses (1.6%, 1/62) and placebo group (0%, 0/35). The secondary end points of major adverse coronary events at 30 and 90 days did not significantly differ but were numerically lower in the PZ-128 groups (0% and 2% in the PZ-128 groups, 6% and 6% with placebo, p=0.13, p=0.29, respectively). In the subgroup of patients with elevated baseline cardiac troponin I, the exploratory end point of 30-day major adverse coronary events + myocardial injury showed 83% events in the placebo group versus 31% events in the combined PZ-128 drug groups, an adjusted relative risk of 0.14 (95% CI, 0.02-0.75); P=0.02. CONCLUSIONS: In this first-in-patient experience, PZ-128 added to standard antiplatelet therapy appeared to be safe, well tolerated, and potentially reduced periprocedural myonecrosis, thus providing the basis for further clinical trials. Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02561000.


Subject(s)
Acute Coronary Syndrome/therapy , Blood Platelets/drug effects , Cardiac Catheterization , Cell-Penetrating Peptides/administration & dosage , Coronary Artery Disease/therapy , Lipopeptides/administration & dosage , Myocardium/pathology , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/administration & dosage , Receptor, PAR-1/agonists , Thrombosis/prevention & control , Acute Coronary Syndrome/diagnostic imaging , Aged , Blood Platelets/metabolism , Cardiac Catheterization/adverse effects , Cardiac Catheterization/instrumentation , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Coronary Artery Disease/diagnostic imaging , Double-Blind Method , Female , Humans , Infusions, Intravenous , Lipopeptides/adverse effects , Lipopeptides/pharmacokinetics , Male , Middle Aged , Necrosis , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/instrumentation , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Proof of Concept Study , Prospective Studies , Receptor, PAR-1/metabolism , Recurrence , Stents , Thrombosis/blood , Thrombosis/etiology , Time Factors , Treatment Outcome , United States
10.
Biochem J ; 477(8): 1363-1366, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32322896

ABSTRACT

Cell-penetrating peptides (CPPs) are short peptides able to cross the cellular membranes without any interaction with specific receptors. Thanks to their ability to transport various cargo inside the cells are emerged as powerful therapeutic agents alternative to small molecules. In recent years, numerous preclinical studies provided promising results for the treatment of various human diseases. Several CPP-conjugated compounds are under clinical trials.


Subject(s)
Cell-Penetrating Peptides/metabolism , Biological Transport , Cell Membrane/metabolism , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Drug Therapy , Humans
11.
Metab Brain Dis ; 36(4): 701-709, 2021 04.
Article in English | MEDLINE | ID: mdl-33420884

ABSTRACT

Alzheimer's disease (AD) is behaviorally characterized by memory impairments, and pathologically by amyloid ß1-42 (Aß1-42) plaques and tangles. Aß binds to excitatory synapses and disrupts their transmission due to dysregulation of the glutamate receptors. Here we hypothesized that chronic inhibition of the endocytosis of AMPA receptors together with GluN2B subunit of NMDA receptors might improve cognition deficit induced by Aß(1-42) neurotoxicity. Forty male Wistar rats were used in this study and divided into 5 groups: Saline + Saline, Aß+Saline, Aß+Ifen (Ifenprodil, 3 nmol /2 weeks), Aß+GluR23Y (Tat-GluR23Y 3 µmol/kg/2 weeks) and Aß+Ifen+GluR23Y (same doses and durations). Aß(1-42) neurotoxicity was induced by intracerebroventricular (ICV) injection of Aß1-42 (2 µg/µl/side), and then animals received the related treatments for 14 days. Cognitive performance of rats and hippocampal level of cAMP-response element-binding (CREB) were evaluated using Morris Water Maze (MWM), and western blotting respectively. Obtained data from the acquisition trials were analyzed by two way Anova and Student T test. Also one way Analysis of variance (ANOVA) with post hoc Tuckey were used to clarify between groups differences in probe test. The Group receiving Aß, showed significant cognition deficit (long latency to platform and short total time spent in target quadrant (TTS), parallel with lower level of hippocampal CREB, versus vehicle group. While, Aß+ GluR23Y exhibited the shortest latency to platform and the longest TTS during the probe test, parallel with the higher hippocampal level of CREB compared with other groups. The present study provides evidence that chronic administration of Tat-GluR23Y; an inhibitor of GluA2-AMPARs endocytosis, successfully restores spatial memory impaired by amyloid beta neurotoxicity targeting CREB signaling pathway.


Subject(s)
Amyloid beta-Peptides/toxicity , Cell-Penetrating Peptides/administration & dosage , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Drug Delivery Systems/methods , Peptide Fragments/toxicity , Animals , Cognitive Dysfunction/chemically induced , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Drug Administration Schedule , Male , Maze Learning/drug effects , Maze Learning/physiology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
12.
Int J Mol Sci ; 22(4)2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33562802

ABSTRACT

Renal ischemia-reperfusion injury (IRI) is involved in the majority of clinical conditions that manifest as renal function deterioration; however, specific treatment for this type of injury is still far from clinical use. Since Toll-like receptor (TLR)-mediated signaling is a key mediator of IRI, we examined the effect of a multiple-TLR-blocking peptide named TLR-inhibitory peptide 1 (TIP1), which exerts the strongest action on TLR4, on renal IRI. We subjected C57BL/6 mice to 23 min of renal pedicle clamping preceded by intraperitoneal injection with a vehicle or TIP1. Sham control mice underwent flank incision only. Mouse kidneys were harvested after 24 h of reperfusion for histology, western blot, RT-PCR, and flow cytometry analysis. Pretreatment with TIP1 lowered the magnitude of elevated plasma creatinine levels and attenuated tubular injury. TIP1 treatment also reduced mRNA expression of inflammatory cytokines and decreased apoptotic cells and oxidative stress in post-ischemic kidneys. In kidneys pretreated with TIP1, the infiltration of macrophages and T helper 17 cells was less abundant than those in the IRI only group. These results suggest that TIP1 has a potential beneficial effect in attenuating the degree of kidney damage induced by IRI.


Subject(s)
Acute Kidney Injury/prevention & control , Cell-Penetrating Peptides/administration & dosage , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Animals , Cell-Penetrating Peptides/pharmacology , Creatinine/blood , Cytokines/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
13.
Int J Mol Sci ; 22(21)2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34769414

ABSTRACT

Cell-penetrating peptides (CPPs) are small peptide sequences used mainly as cellular delivery agents that are able to efficiently deliver cargo into cells. Some CPPs also demonstrate intrinsic anticancer properties. Previously, our group developed a new family of CPP2-thiazole conjugates that have been shown to effectively reduce the proliferation of different cancer cells. This work aimed to combine these CPP2-thiazole conjugates with paclitaxel (PTX) and 5-fluorouracil (5-FU) in PC-3 prostate and HT-29 colon cancer cells, respectively, to evaluate the cytotoxic effects of these combinations. We also combined these CPP2-thiazole conjugates with clotrimazole (CLZ), an antifungal agent that has been shown to decrease cancer cell proliferation. Cell viability was evaluated using MTT and SRB assays. Drug interaction was quantified using the Chou-Talalay method. We determined that CPP2 did not have significant activity in these cells and demonstrate that N-terminal modification of this peptide enhanced its anticancer activity in both cell lines. Our results also showed an uneven response between cell lines to the proposed combinations. PC-3 cells were more responsive to the combination of CPP2-thiazole conjugates with CLZ than PTX and were more sensitive to these combinations than HT-29 cells. In addition, the interaction of drugs resulted in more synergism in PC-3 cells. These results suggest that N-terminal modification of CPP2 results in the enhanced anticancer activity of the peptide and demonstrates the potential of CPPs as adjuvants in cancer therapy. These results also validate that CLZ has significant anticancer activity both alone and in combination and support the strategy of drug repurposing coupled to drug combination for prostate cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell-Penetrating Peptides/pharmacology , Clotrimazole/pharmacology , Colonic Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Thiazoles/pharmacology , Antifungal Agents/administration & dosage , Antifungal Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Clotrimazole/administration & dosage , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Synergism , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Humans , Male , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Thiazoles/administration & dosage , Thiazoles/chemistry
14.
Molecules ; 26(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805680

ABSTRACT

This review summarizes recent developments in conjugation techniques for the synthesis of cell-penetrating peptide (CPP)-drug conjugates targeting cancer cells. We will focus on small organic molecules as well as metal complexes that were used as cytostatic payloads. Moreover, two principle ways of coupling chemistry will be discussed direct conjugation as well as the use of bifunctional linkers. While direct conjugation of the drug to the CPP is still popular, the use of bifunctional linkers seems to gain increasing attention as it offers more advantages related to the linker chemistry. Thus, three main categories of linkers will be highlighted, forming either disulfide acid-sensitive or stimuli-sensitive bonds. All techniques will be thoroughly discussed by their pros and cons with the aim to help the reader in the choice of the optimal conjugation technique that might be used for the synthesis of a given CPP-drug conjugate.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemical synthesis , Cytostatic Agents/administration & dosage , Cytostatic Agents/chemical synthesis , Drug Delivery Systems/methods , Amino Acid Sequence , Animals , Cell Line, Tumor , Cytostatic Agents/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemical synthesis , Drug Carriers/chemistry , Drug Delivery Systems/trends , Humans , Molecular Structure , Organic Chemistry Phenomena
15.
Pharm Dev Technol ; 26(6): 634-646, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33843423

ABSTRACT

A combination of doxorubicin (DOX) and small interfering RNA (siRNA) is proven effective for the reverse of multidrug resistance. However, rapid degradation and poor cellular internalization of siRNA hinder their synergistic action. To improve the combination effect, asparagine-glycine-arginine peptide (NGR) -modified nanobubbles (NBs) containing cell-penetrating peptide (CPP) decorated DOX and CPP decorated c-myc siRNA were constructed. Diameters of these NBs were about 245 nm and zeta potentials were about -3 mV. Encapsulation efficiencies (EE) of DOX exceeded 80%. Release of DOX could be triggered by ultrasound (US) since above 80% DOX was released from NBs after sonication while less than 5% DOX was discharged without treatment of US. These NBs were considered stable during 24 h since the decrease of particle size was no more than 10 nm, variances of EE were less than 5%, and changes of transmission (ΔT) were less than 3%. More drugs in formulation decorated with CPP and NGR were accumulated in the tumor when combined with sonication. The evident synergistic action of DOX, siRNA, NBs, and US was verified in mice with strong antitumor efficacy. Taken together, NGR-modified NBs containing CPP-DOX and CPP-siRNA are able to realize time- and spatial-controlled drug delivery and show potential application prospects.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Nanoparticles/administration & dosage , Oligopeptides/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Antibiotics, Antineoplastic/administration & dosage , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , RNA, Small Interfering/genetics , Tumor Burden/drug effects , Tumor Burden/physiology
16.
Biochem Biophys Res Commun ; 533(4): 905-912, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33008588

ABSTRACT

Cell-penetrating peptides (CPPs) can deliver payloads into cells by forming complexes with bioactive molecules via either covalent or non-covalent bonds. Previously, we reported polyhistidine (H16 peptide: HHHHHHHHHHHHHHHH-NH2) as a new CPP. This peptide is anticipated to be a valuable new carrier for drug delivery to intracellular lysosomes; the peptide can transport macromolecules into these organelles. In the present study, we examined the application of the H16 peptide as a drug delivery system (DDS) to reverse to lysosomal storage disease (LSD) in cells in vitro. LSDs are metabolic disorders caused by the loss of specific lysosomal enzymes. The majority of lysosomal enzymes are acidic proteins and we utilized this common feature for our DDS. We synthesized a polylysine-polyhistidine fusion peptide (K10H16 peptide: KKKKKKKKKKGHHHHHHHHHHHHHHHH-NH2) and developed a simple method for transporting acidic proteins into intracellular lysosomes via formation of complexes of enzymes with the K10H16 peptide by electrostatic interaction. First, we demonstrated our strategy using maltose-binding protein-fused green fluorescent protein (MBP-GFP) to model an acidic protein. The K10H16 peptide bound to MBP-GFP and transported it into intracellular lysosomes. Further, alpha-galactosidase A (GLA), one of the lysosomal enzymes associated with LSD, was also delivered to intracellular lysosomes by the peptide. The complex between K10H16 peptide and GLA restored typical proliferation to LSD cells, which otherwise grew more slowly than normal cells. These results suggest that K10H16 peptide replenished lysosomal enzyme deficiency in LSD cells. The K10H16 peptide may be useful as a DDS for LSD therapy.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Drug Delivery Systems , Enzyme Replacement Therapy/methods , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/enzymology , Lysosomes/drug effects , Lysosomes/metabolism , Amino Acid Sequence , Cells, Cultured , Fabry Disease/drug therapy , Fabry Disease/enzymology , Histidine/chemistry , Humans , Polylysine/chemistry , Protein Transport , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , alpha-Galactosidase/administration & dosage
17.
J Pharmacol Exp Ther ; 374(3): 354-365, 2020 09.
Article in English | MEDLINE | ID: mdl-32561686

ABSTRACT

The development of neuropharmaceutical gene delivery systems requires strategies to obtain efficient and effective brain targeting as well as blood-brain barrier (BBB) permeability. A brain-targeted gene delivery system based on a transferrin (Tf) and cell-penetrating peptide (CPP) dual-functionalized liposome, CPP-Tf-liposome, was designed and investigated for crossing BBB and permeating into the brain. We selected three sequences of CPPs [melittin, Kaposi fibroblast growth factor (kFGF), and penetration accelerating sequence-R8] and compared their ability to internalize into the cells and, subsequently, improve the transfection efficiency. Study of intracellular uptake indicated that liposomal penetration into bEnd.3 cells, primary astrocytes, and primary neurons occurred through multiple endocytosis pathways and surface modification with Tf and CPP enhanced the transfection efficiency of the nanoparticles. A coculture in vitro BBB model reproducing the in vivo anatomophysiological complexity of the biologic barrier was developed to characterize the penetrating properties of these designed liposomes. The dual-functionalized liposomes effectively crossed the in vitro barrier model followed by transfecting primary neurons. Liposome tissue distribution in vivo indicated superior ability of kFGF-Tf-liposomes to overcome BBB and reach brain of the mice after single intravenous administration. These findings demonstrate the feasibility of using strategically designed liposomes by combining Tf receptor targeting with enhanced cell penetration as a potential brain gene delivery vector. SIGNIFICANCE STATEMENT: Rational synthesis of efficient brain-targeted gene carrier included modification of liposomes with a target-specific ligand, transferrin, and with cell-penetrating peptide to enhance cellular internalization. Our study used an in vitro triple coculture blood-brain barrier (BBB) model as a tool to characterize the permeability across BBB and functionality of designed liposomes prior to in vivo biodistribution studies. Our study demonstrated that rational design and characterization of BBB permeability are efficient strategies for development of brain-targeted gene carriers.


Subject(s)
Brain/drug effects , Liposomes/administration & dosage , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Cell-Penetrating Peptides/administration & dosage , Drug Delivery Systems/methods , Female , Gene Transfer Techniques , Genetic Therapy/methods , Male , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Neurons/drug effects , Neurons/metabolism , Rats, Sprague-Dawley , Tissue Distribution/physiology , Transferrin/administration & dosage
18.
Biomacromolecules ; 21(2): 472-483, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31756087

ABSTRACT

A series of amino- and guanidino-terminating 3- and 4-generation 2,4-diaminobutanoic acid (Dab) dendrons have been robustly synthesized on a solid phase and characterized as cellular delivery agents in antisense peptide nucleic acid (PNA) conjugates in the pLuc705 HeLa cell splice switching system. The dendron-PNA conjugates exhibited splice correction activity at one digit micromolar concentrations, and guanidino-terminating dendrons were significantly more effective than analogous amine terminating ones. Furthermore, introduction of lipophilic groups such as phenyl, alkyl, or fatty acids increased efficacy, but also increased cellular toxicity. Fluorescence microscopy analyses supported an endosomal uptake mechanism and furthermore predominantly showed colocalization with late endosomes and lysosomes. The robust solid phase synthesis should make such Dab-dendrons a useful platform for further in vitro as well as in vivo optimization.


Subject(s)
Dendrimers/chemistry , Peptide Nucleic Acids/administration & dosage , Peptide Nucleic Acids/chemistry , Aminobutyrates/chemistry , Cell-Penetrating Peptides/administration & dosage , HeLa Cells , Humans , Luciferases/genetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Peptide Nucleic Acids/pharmacology , Solid-Phase Synthesis Techniques/methods , Structure-Activity Relationship
19.
Pharmacol Res ; 162: 105231, 2020 12.
Article in English | MEDLINE | ID: mdl-33027717

ABSTRACT

Cancer is the second leading cause of death in the world and its treatment is extremely challenging, mainly due to its complexity. Cell-Penetrating Peptides (CPPs) are peptides that can transport into the cell a wide variety of biologically active conjugates (or cargoes), and are, therefore, promising in the treatment and in the diagnosis of several types of cancer. Some notable examples are TAT and Penetratin, capable of penetrating the central nervous system (CNS) and, therefore, acting in cancers of this system, such as Glioblastoma Multiforme (GBM). These above-mentioned peptides, conjugated with traditional chemotherapeutic such as Doxorubicin (DOX) and Paclitaxel (PTX), have also been shown to induce apoptosis of breast and liver cancer cells, as well as in lung cancer cells, respectively. In other cancers, such as esophageal cancer, the attachment of Magainin 2 (MG2) to Bombesin (MG2B), another CPP, led to pronounced anticancer effects. Other examples are CopA3, that selectively decreased the viability of gastric cancer cells, and the CPP p28. Furthermore, in preclinical tests, the anti-tumor efficacy of this peptide was evaluated on human breast cancer, prostate cancer, ovarian cancer, and melanoma cells in vitro, leading to high expression of p53 and promoting cell cycle arrest. Despite the numerous in vitro and in vivo studies with promising results, and the increasing number of clinical trials using CPPs, few treatments reach the expected clinical efficacy. Usually, their clinical application is limited by its poor aqueous solubility, immunogenicity issues and dose-limiting toxicity. This review describes the most recent advances and innovations in the use of CPPs in several types of cancer, highlighting their crucial importance for various purposes, from therapeutic to diagnosis. Further clinical trials with these peptides are warranted to examine its effects on various types of cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell-Penetrating Peptides/administration & dosage , Drug Delivery Systems , Neoplasms/drug therapy , Animals , Humans
20.
Soft Matter ; 16(20): 4746-4755, 2020 May 28.
Article in English | MEDLINE | ID: mdl-32329496

ABSTRACT

Penetratin is a short Trojan peptide that attracts great interest in biomedical research for its capacity to translocate biological membranes. Herein, we study in detail both self-assembly and intracellular delivery of DNA by the heptamer KIWFQNR, a truncated peptide derived from Penetratin. This shortened sequence possesses a unique design with bolaamphiphilic characteristics that preserves the longest noncationic amino acid portion found in Penetratin. These features convey amphipathicity to assist self-assembly and make it a suitable model for exploring the role of hydrophobic residues for peptide interaction and cell uptake. We show that the fragment forms peptiplexes (i.e., peptide-DNA complexes), and aggregates into long nanofibers with clear ß-sheet signature. The supramolecular structure of nanofibers is likely composed of DNA cores surrounded by a peptide shell to which the double helix behaves as a template and induces fibrillization. A nucleation and growth mechanism proceeding through liquid-liquid phase separation of coacervates is proposed for describing the self-assembly of peptiplexes. We also demonstrate that peptiplexes deliver double-stranded 200 bp DNA into HeLa cells, indicating its potential for preparing non-viral vectors for oligonucleotides through noncovalent strategies. Since the main structural features of native Penetratin are conserved in this simpler fragment, our findings also highlight the role of uncharged amino acids for structuration, and thus for the ability of Penetratin to cross cell membranes.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , DNA/administration & dosage , Peptide Fragments/administration & dosage , Cell-Penetrating Peptides/chemistry , Cytosol/metabolism , DNA/chemistry , Endocytosis , HeLa Cells , Humans , Models, Molecular , Peptide Fragments/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL