Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
ACS Chem Biol ; 16(2): 414-428, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33533253

ABSTRACT

Peptides are being developed as targeted anticancer drugs to modulate cytosolic protein-protein interactions involved in cancer progression. However, their use as therapeutics is often limited by their low cell membrane permeation and/or inability to reach cytosolic targets. Conjugation to cell penetrating peptides has been successfully used to improve the cytosolic delivery of high affinity binder peptides, but cellular uptake does not always result in modulation of the targeted pathway. To overcome this limitation, we developed "angler peptides" by conjugating KD3, a noncell permeable but potent and specific peptide inhibitor of p53:MDM2 and p53:MDMX interactions, with a set of cyclic cell-penetrating peptides. We examined their binding affinity for MDM2 and MDMX, the cell entry mechanism, and role in reactivation of the p53 pathway. We identified two angler peptides, cTAT-KD3 and cR10-KD3, able to activate the p53 pathway in cancer cells. cTAT-KD3 entered cells via endocytic pathways, escaped endosomes, and activated the p53 pathway in breast (MCF7), lung (A549), and colon (HCT116) cancer cell lines at concentrations in the range of 1-12 µM. cR10-KD3 reached the cytosol via direct membrane translocation and activated the p53 pathway at 1 µM in all the tested cell lines. Our work demonstrates that nonpermeable anticancer peptides can be delivered into the cytosol and inhibit intracellular cancer pathways when they are conjugated with stable cell penetrating peptides. The mechanistic studies suggest that direct translocation leads to less toxicity, higher cytosol delivery at lower concentrations, and lower dependencies on the membrane of the tested cell line than occurs for an endocytic pathway with endosomal escape. The angler strategy can rescue high affinity peptide binders identified from high throughput screening and convert them into targeted anticancer therapeutics, but investigation of their cellular uptake and cell death mechanisms is essential to confirming modulation of the targeted cancer pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Peptides, Cyclic/pharmacology , Protein Binding/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/toxicity , Drug Design , Drug Screening Assays, Antitumor , Erythrocytes , Humans , Leukocytes, Mononuclear/drug effects , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/toxicity , Protein Conformation, alpha-Helical
2.
ACS Appl Mater Interfaces ; 12(51): 56815-56829, 2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33296603

ABSTRACT

Anticancer lipopeptides (ACLPs) are considered promising alternatives to combat resistant cancer cells, but the influence of peptide conformational propensity alone on their selectivity and mechanism remains obscure. In this study, we developed N-palmitoylated MK5E (P1MK5E) and MEK5 (P1MEK5) that have the same composition of 23 residues undergoing the pH-dependent structural alterations but differ in the conformational tendency of their amino acid composites. Nonlipidated peptides were readily accumulated in the A549 cell nucleus by the direct membrane translocation and the heparan sulfate-mediated endocytosis than the lipid-raft-dependent pathway. The increased hydrophobicity favored the amino acid-position-dependent folding of P1MK5E and P1MEK5, respectively, toward the α-helical coiled-coil nanofibrils and amyloidlike ß-protofibrils. At the close concentrations (∼7.5 µM) to the toxic effects of doxorubicin (DOX), P1MK5E exhibited (i) an increased anticancer toxicity through a time-dependent S-phase arrest, (ii) enhanced plasma membrane permeability, and (iii) dose-dependent changes in the cell death characteristic features in the A549 cells relative to P1MEK5 that was almost inactive at ∼75 µM. These observations were in accordance with the TNF-α-mediated necroptotic signaling in the c-MYC/PARP1-overexpressed A549 cells exposed to P1MK5E and accompanied by the ultrastructure of plasma membrane protrusions, extensive endoplasmic reticulum (ER) membrane expansion, mitochondrial swelling, and the formation of distinct cytoplasmic vacuolation. The structural results and the bioactivity behaviors, herein, declared the significance of α-helical propensity in the peptide sequence and the nanostructure morphologies of self-assembling ACLPs upon the selectivity and enhanced anticancer effectiveness, which notably holds promise in the design and development of efficient therapeutics for cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Magainins/pharmacology , Necroptosis/drug effects , A549 Cells , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , CHO Cells , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/toxicity , Cricetulus , Drug Screening Assays, Antitumor , Human Umbilical Vein Endothelial Cells , Humans , Hydrophobic and Hydrophilic Interactions , Lipoylation , Magainins/chemistry , Magainins/toxicity , Protein Structure, Secondary , S Phase Cell Cycle Checkpoints/drug effects
3.
Acta Biomater ; 109: 195-207, 2020 06.
Article in English | MEDLINE | ID: mdl-32294553

ABSTRACT

Stem cell therapy holds great promise for cardiac regeneration. However, the lack of ability to control stem cell fate after in vivo transplantation greatly restricts its therapeutic outcomes. MicroRNA delivery has emerged as a powerful tool to control stem cell fate for enhanced cardiac regeneration. However, the clinical translation of therapy based on gene-transfected stem cells remains challenging, due to the unknown in vivo behaviors of stem cells. Here, we developed a nano-platform (i.e., PFBT@miR-1-Tat NPs) that can achieve triggered release of microRNA-1 to promote cardiac differentiation of mesenchymal stem cells (MSCs), and long-term tracking of transplanted MSCs through bright and ultra-stable fluorescence of conjugated polymer poly(9,9-dioctylfluorene-alt-benzothiadiazole) (PFBT). We found that PFBT@miR-1-Tat NP-treated MSCs significantly restored the infarcted myocardium by promoting stem cell cardiac differentiation and integration with the in situ cardiac tissues. Meanwhile, MSCs without gene delivery improved the infarcted heart functions mainly through a paracrine effect and blood vessel formation. The developed conjugated polymer nanovector should be a powerful tool for manipulating as well as revealing the fate of therapeutic cells in vivo, which is critical for optimizing the therapeutic route of gene and cell combined therapy and therefore for accelerating clinical translation. STATEMENT OF SIGNIFICANCE: The lack of controllability in stem cell fate and the unclear in vivo cellular behaviors restrict the therapeutic outcomes of stem cell therapy. Herein, we engineered fluorescent conjugated polymer nanoparticles as gene delivery nanovectors with controlled release and high intracellular delivery capability to harness the fate of mesenchymal stem cells (MSCs) in vivo, meanwhile to reveal the cellular mechanism of gene-treated stem cell therapy. As compared with only MSC treatment that improves infarcted myocardium functions through paracrine effect, treatment with conjugated polymer nanovector-treated MSCs significantly restored infarcted myocardium through enhancing MSC cardiac differentiation and integration with the in-situ cardiac tissues. These findings demonstrate that the conjugated polymer nanovector would be a powerful tool in optimizing gene and cell combined therapy.


Subject(s)
Drug Carriers/chemistry , Mesenchymal Stem Cells/metabolism , MicroRNAs/therapeutic use , Myocardial Infarction/therapy , Nanoparticles/chemistry , Animals , Cell Differentiation/drug effects , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/toxicity , Drug Carriers/toxicity , Fluorenes/chemistry , Fluorenes/toxicity , Heart/physiology , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , MicroRNAs/chemistry , Myocardium/pathology , Myocytes, Cardiac/metabolism , Nanoparticles/toxicity , Polymers/chemistry , Polymers/toxicity , Rats, Sprague-Dawley , Regeneration
4.
Nat Commun ; 11(1): 1733, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32265441

ABSTRACT

Dysregulation of extracellular signal-regulated kinases (ERK1/2) is linked to several diseases including heart failure, genetic syndromes and cancer. Inhibition of ERK1/2, however, can cause severe cardiac side-effects, precluding its wide therapeutic application. ERKT188-autophosphorylation was identified to cause pathological cardiac hypertrophy. Here we report that interference with ERK-dimerization, a prerequisite for ERKT188-phosphorylation, minimizes cardiac hypertrophy without inducing cardiac adverse effects: an ERK-dimerization inhibitory peptide (EDI) prevents ERKT188-phosphorylation, nuclear ERK1/2-signaling and cardiomyocyte hypertrophy, protecting from pressure-overload-induced heart failure in mice whilst preserving ERK1/2-activity and cytosolic survival signaling. We also examine this alternative ERK1/2-targeting strategy in cancer: indeed, ERKT188-phosphorylation is strongly upregulated in cancer and EDI efficiently suppresses cancer cell proliferation without causing cardiotoxicity. This powerful cardio-safe strategy of interfering with ERK-dimerization thus combats pathological ERK1/2-signaling in heart and cancer, and may potentially expand therapeutic options for ERK1/2-related diseases, such as heart failure and genetic syndromes.


Subject(s)
Cardiotoxicity , Cell-Penetrating Peptides/pharmacology , Dimerization , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Animals , Cell Culture Techniques , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/toxicity , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Drug Delivery Systems , Extracellular Signal-Regulated MAP Kinases/drug effects , Heart Failure/drug therapy , Heart Failure/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Molecular Medicine , Rats , Rats, Sprague-Dawley , Signal Transduction
5.
Org Biomol Chem ; 18(10): 1978-1986, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32104826

ABSTRACT

Development of an intracellular delivery method for functional peptides via cell-penetrating peptides (CPPs) expands peptide use in basic research and therapeutic applications. Although direct conjugation of a functional peptide with a CPP is the simplest method for delivery, this method has not always been reliable. CPPs usually contain several positively charged amino acids that potentially interact non-specifically with negatively charged molecules in cells and subsequently interfere with conjugated functional peptide function. Here we demonstrate a new intracellular delivery method for peptides in which a functional peptide is released from a positively charged CPP via peptide nucleic acids (PNAs). We prepared an 8-mer PNA conjugated to octa-arginine in tandem (PNA1-CPP) and linked its complementary PNA to an autophagy inducing peptide (PNA2-AIP) by solid-phase peptide synthesis. PNA1-CPP and PNA2-AIP formed a 1 : 1 hybrid via PNA1/PNA2 interaction, thereby indirectly but stably connecting the AIP to the CPP. PNA2-AIP was successfully delivered into cells in a hybrid formation-dependent manner and at least some portion of the PNA1-CPP/PNA2-AIP hybrids dissociated into PNA2-AIP and PNA1-CPP inside the cells. Notably, PNA2-AIP delivered to cells induced more autophagy than AIP directly conjugated to CPP (CPP-AIP). Further, the PNA hybrid did not induce significant cell death. These findings indicate that the PNA1/PNA2 hybrid can function as a molecular glue enabling the delivery of functional peptides into cells.


Subject(s)
Beclin-1/pharmacology , Cell-Penetrating Peptides/metabolism , Drug Carriers/metabolism , Peptide Fragments/pharmacology , Peptide Nucleic Acids/metabolism , Autophagy/drug effects , Beclin-1/metabolism , Beclin-1/toxicity , Cell-Penetrating Peptides/toxicity , Drug Carriers/toxicity , Drug Liberation , HeLa Cells , Humans , Leucine Zippers , Oligopeptides/metabolism , Oligopeptides/toxicity , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Peptide Nucleic Acids/toxicity , Protein Binding
6.
Chem Commun (Camb) ; 56(1): 145-148, 2019 Dec 17.
Article in English | MEDLINE | ID: mdl-31799976

ABSTRACT

The selective measurement of cellular pH at specific regions is significant for the in-depth study of cell functions and pathological processes. Herein, we combined our previously developed rhodamine B-labeled cell-penetrating peptide spirolactam derivative with quantum dots to create a ratiometric nanobiosensor for the simultaneous sensing and imaging of lysosomes and extracellular pH.


Subject(s)
Cell-Penetrating Peptides/chemistry , Lysosomes/metabolism , Quantum Dots/chemistry , Biosensing Techniques/methods , Cadmium Compounds/chemistry , Cadmium Compounds/toxicity , Cell-Penetrating Peptides/toxicity , Fluorescent Dyes/chemistry , Fluorescent Dyes/toxicity , HeLa Cells , Humans , Hydrogen-Ion Concentration , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Nanoconjugates/chemistry , Nanoconjugates/toxicity , Quantum Dots/toxicity , Rhodamines/chemistry , Rhodamines/toxicity , Selenium Compounds/chemistry , Selenium Compounds/toxicity , Sulfides/chemistry , Sulfides/toxicity , Zinc Compounds/chemistry , Zinc Compounds/toxicity
7.
Expert Opin Drug Deliv ; 16(11): 1227-1258, 2019 11.
Article in English | MEDLINE | ID: mdl-31583914

ABSTRACT

Introduction: Cell penetrating peptides (CPPs) known as protein translocation domains (PTD), membrane translocating sequences (MTS), or Trojan peptides (TP) are able to cross biological membranes without clear toxicity using different mechanisms, and facilitate the intracellular delivery of a variety of bioactive cargos. CPPs could overcome some limitations of drug delivery and combat resistant strains against a broad range of diseases. Despite delivery of different therapeutic molecules by CPPs, they lack cell specificity and have a short duration of action. These limitations led to design of combined cargo delivery systems and subsequently improvement of their clinical applications. Areas covered: This review covers all our studies and other researchers in different aspects of CPPs such as classification, uptake mechanisms, and biomedical applications. Expert opinion: Due to low cytotoxicity of CPPs as compared to other carriers and final degradation to amino acids, they are suitable for preclinical and clinical studies. Generally, the efficiency of CPPs was suitable to penetrate the cell membrane and deliver different cargos to specific intracellular sites. However, no CPP-based therapeutic approach has approved by FDA, yet; because there are some disadvantages for CPPs including short half-life in blood, and nonspecific CPP-mediated delivery to normal tissue. Thus, some methods were used to develop the functions of CPPs in vitro and in vivo including the augmentation of cell specificity by activatable CPPs, specific transport into cell organelles by insertion of corresponding localization sequences, incorporation of CPPs into multifunctional dendrimeric or liposomal nanocarriers to improve selectivity and efficiency especially in tumor cells.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Drug Carriers/administration & dosage , Animals , Biological Transport , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/toxicity , Drug Carriers/chemistry , Drug Carriers/toxicity , Humans , Structure-Activity Relationship
8.
Analyst ; 144(21): 6254-6261, 2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31560359

ABSTRACT

As VEGF mRNA is an endothelial cell-specific mitogen and a key regulator of angiogenesis in a variety of physiological and pathological processes, high expression levels of VEGF messenger RNA (mRNA) contribute to VEGF-driven angiogenesis in the hypoxic areas of solid tumors and then disrupt the vascular barrier, which may potentiate tumor cell extravasation. Thus, monitoring the changes in VEGF mRNA is necessary to understand the genetic programme under hypoxic conditions and thus facilitate risk assessment and risk reduction in hypoxic environments. Herein, a new fluorescent nanoprobe based on azoreductase-responsive functional metal-organic frameworks (AMOFs) was developed to realize the imaging of VEGF mRNA under hypoxic conditions. Since the azobenzene units in the AMOFs can be reduced to amines by the highly expressed azoreductase in an oxygen-deficient environment, the VEGF mRNA-targeted molecular beacon (MB), which is adsorbed on the surface of AMOFs via electrostatic interactions, can be released due to the structural damage of AMOFs. Moreover, TAMRA (carboxytetramethylrhodamine, donor) and Cy5 (acceptor) were close to each other due to the stem-loop conformation of MB, thus inducing high fluorescence energy resonance transfer (FRET) efficiency. Upon the addition of VEGF mRNA, the hybridization of VEGF mRNA destroyed the stem-loop conformation of MB, and then, the two fluorophores labeled on MB were separated with low FRET efficiency. This constructed fluorescent nanoprobe enables the quantitative analysis and in situ imaging of the VEGF mRNA level in living cells under hypoxic conditions. We expect that it will offer a potentially rich opportunity to understand the physiological processes of genetic programme.


Subject(s)
Cell Hypoxia/physiology , Fluorescent Dyes/chemistry , Metal-Organic Frameworks/chemistry , NADH, NADPH Oxidoreductases/chemistry , RNA, Messenger/analysis , Vascular Endothelial Growth Factor A/genetics , Carbocyanines/chemistry , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/toxicity , DNA/chemistry , DNA/genetics , DNA Probes/chemistry , DNA Probes/genetics , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/toxicity , HeLa Cells , Humans , Metal-Organic Frameworks/chemical synthesis , Metal-Organic Frameworks/toxicity , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Nitroreductases , Nucleic Acid Hybridization , RNA, Messenger/genetics , Rhodamines/chemistry
9.
ACS Chem Biol ; 14(9): 2071-2087, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31390185

ABSTRACT

The tumor suppressor protein p53 is inactive in a large number of cancers, including some forms of sarcoma, breast cancer, and leukemia, due to overexpression of its intrinsic inhibitors MDM2 and MDMX. Reactivation of p53 tumor suppressor activity, via disruption of interactions between MDM2/X and p53 in the cytosol, is a promising strategy to treat cancer. Peptides able to bind MDM2 and/or MDMX were shown to prevent MDM2/X:p53 interactions, but most possess low cell penetrability, low stability, and/or high toxicity to healthy cells. Recently, the designed peptide cHLH-p53-R was reported to possess high affinity for MDM2, resistance toward proteases, cell-penetrating properties, and toxicity toward cancer cells. This peptide uses a stable cyclic helix-loop-helix (cHLH) scaffold, which includes two helices connected with a Gly loop and cyclized to improve stability. In the current study, we were interested in examining the cell selectivity of cHLH-p53-R, its cellular internalization, and ability to reactivate the p53 pathway. We designed analogues of cHLH-p53-R and employed biochemical and biophysical methodologies using in vitro model membranes and cell-based assays to compare their structure, activity, and mode-of-action. Our studies show that cHLH is an excellent scaffold to stabilize and constrain p53-mimetic peptides with helical conformation, and reveal that anticancer properties of cHLH-p53-R are mediated by its ability to selectively target, cross, and disrupt cancer cell membranes, and not by activation of the p53 pathway. These findings highlight the importance of examining the mode-of-action of designed peptides to fully exploit their potential to develop targeted therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane/metabolism , Cell-Penetrating Peptides/pharmacology , Peptides, Cyclic/pharmacology , Tumor Suppressor Proteins/pharmacology , Amino Acid Sequence , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/toxicity , Helix-Loop-Helix Motifs , Humans , Lipid Bilayers/metabolism , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/toxicity , Protein Binding/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/chemical synthesis , Tumor Suppressor Proteins/toxicity
10.
Microb Pathog ; 136: 103689, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31445122

ABSTRACT

As an intracellular pathogen, Listeria monocytogenes can enter host cells where it can replicate and escape detection and eradication by the host immune response making the clearance of infection very challenging. Furthermore, with the advent of antimicrobial resistance, the need for alternative targets is inevitable. Internalin proteins are crucial to this bacterium as they contribute to bacterial entry to the systemic circulation. In this study, we targeted a highly conserved region of these proteins by an antisense sequence that was covalently conjugated to the cell penetrating peptides (CPP) to overcome the challenging delivery barriers. Then, we evaluated the efficiency of this construct in vitro. We also assessed the antigenicity, cytotoxicity, and probability of apoptosis induction by this construct. The studied CPP-PNA inhibited bacterial growth and suppressed the mRNA expression of internalins in a dose-dependent manner. In addition, at all studied concentrations, CPP-PNA significantly reduced the invasion rate of L. monocytogenes in the examined cell lines. Moreover, different concentrations of CPP-PNA did not have a significant antigenic, cytotoxic, and apoptotic properties compared to the control. These results suggest the effectiveness of CPP-antisense in targeting the mRNAs of internalins for various research, therapeutic and preventive purposes. However, additional research is required to evaluate the potency, safety, and pharmacokinetics of this compound for the prevention and treatment of listeriosis.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Proteins/antagonists & inhibitors , Endocytosis/drug effects , Listeria monocytogenes/drug effects , Listeriosis/prevention & control , Oligodeoxyribonucleotides, Antisense/metabolism , Anti-Bacterial Agents/toxicity , Bacterial Proteins/genetics , Caco-2 Cells , Cell Survival/drug effects , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/toxicity , Consensus Sequence , HeLa Cells , Humans , Listeria monocytogenes/genetics , Microbial Sensitivity Tests , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/toxicity
11.
ChemMedChem ; 14(16): 1547-1559, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31359624

ABSTRACT

Phenol-soluble modulins (PSMs) are a large family of cytolytic peptide toxins produced by Staphylococcus aureus. Based on their amino acid sequences, we have constructed a small library of cationic isoleucine-rich peptides for antimicrobial evaluation. Relative to the parent PSMs, peptide zp3 (GIIAGIIIKIKK-NH2 ) was found to possess greatly improved physicochemical properties (soluble in water) and antibacterial activity (MIC=8 µm for E. coli, B. subtilis, and C. freundii) while maintaining low hemolytic activity (<5 % at 256 µm) and cytotoxicity (HEK293 cells IC50 >80 µm). We reasoned that the selective activity of zp3 toward bacterial cells is due to its amphiphilic nature and positive net charge. Moreover, it is difficult for bacteria to develop resistance against zp3. Through microscopic studies of E. coli, we demonstrated that zp3 can penetrate the bacterial membrane, thereby causing leakage of the bacterial cytoplasm. Our findings present a promising antimicrobial peptide lead, which has great potential for further chemical modification.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Surface-Active Agents/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Bacteria/drug effects , Cell Membrane/drug effects , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/toxicity , Drug Design , Erythrocytes/drug effects , HEK293 Cells , Hemolysis/drug effects , Humans , Microbial Sensitivity Tests , Peptide Library , Rats , Surface-Active Agents/chemistry , Surface-Active Agents/toxicity
12.
Chem Commun (Camb) ; 55(54): 7792-7795, 2019 Jul 02.
Article in English | MEDLINE | ID: mdl-31210205

ABSTRACT

We have designed and synthesized a set of cell-penetrating foldamers (CPFs), Blocks 1-8, composed of the common amino acids Leu, Arg, and Gly, as well as the helicogenic amino acid 2-aminoisobutyric acid. The findings showed that Block 3 could deliver siRNA into cells without significant cytotoxicity. We also demonstrated that Block 3 could be applied to selectively kill the oncogene-driven cancer cells.


Subject(s)
Aminoisobutyric Acids/chemistry , Cell-Penetrating Peptides/metabolism , Drug Carriers/metabolism , RNA, Small Interfering/administration & dosage , Amino Acid Sequence , Cell Line, Tumor , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/toxicity , Drug Carriers/chemical synthesis , Drug Carriers/chemistry , Drug Carriers/toxicity , Gene Knockdown Techniques , Gene Silencing , Humans , Protein Conformation, alpha-Helical , Protein Engineering/methods , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Small Interfering/genetics
13.
Exp Biol Med (Maywood) ; 244(8): 621-629, 2019 05.
Article in English | MEDLINE | ID: mdl-30836793

ABSTRACT

IMPACT STATEMENT: Bax induces mitochondria-dependent programed cell death. While cytotoxic drugs activating Bax have been developed for cancer treatment, clinically effective therapeutics suppressing Bax-induced cell death rescuing essential cells have not been developed. This mini-review will summarize previously reported Bax inhibitors including peptides, small compounds, and antibodies. We will discuss potential applications and the future direction of these Bax inhibitors.


Subject(s)
Apoptosis/drug effects , Cell-Penetrating Peptides/pharmacology , bcl-2-Associated X Protein/antagonists & inhibitors , Amino Acid Sequence , Animals , Apoptosis/physiology , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/therapeutic use , Cell-Penetrating Peptides/toxicity , Cells, Cultured , Disease Models, Animal , Drug Carriers , Drug Design , Humans , Immunoglobulin Fab Fragments/pharmacology , Ku Autoantigen/metabolism , Mice , Mice, Knockout , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Permeability Transition Pore , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Organ Preservation/methods , Pinocytosis , Protein Multimerization/drug effects , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Rats , Retinal Degeneration/drug therapy , Retinal Degeneration/pathology , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/immunology , bcl-2-Associated X Protein/metabolism
14.
Int J Nanomedicine ; 14: 605-622, 2019.
Article in English | MEDLINE | ID: mdl-30697045

ABSTRACT

BACKGROUND: Fungal keratitis is a major cause of corneal blindness accounting for more than one-third of microbiologically proven cases. The management of fungal keratitis is through topical or systemic antifungal medications alone or in combination with surgical treatment. Topical medications such as natamycin and voriconazole pose major challenges due to poor penetration across the corneal epithelium. To address the issue various carrier molecules like nanoparticles, lipid vesicles, and cell penetrating peptides were explored. But the major drawback such as non-specificity and lack of bioavailability remains. PURPOSE: In this study, we have attempted to design corneal specific cell penetrating peptide using subtractive proteomic approach from the published literature and tried to improve its bioavailability through gelatin hydrogel delivery system. MATERIAL AND METHODS: Using subtractive proteomic approach two peptides VRF005 and VRF007 were identified on the basis of solubility, cell permeability and amphipathicity. The peptides were modeled for three-dimensional structure and simulated for membrane penetration. The peptides were characterized using circular dichroism spectroscopy, dynamic light scattering and native polyacrylamide gel electrophoresis. Further uptake studies were performed on primary corneal epithelial cells and the stability was analyzed in corneal epithelial tissue lysates. Insilico prediction of peptides showed it to have antifungal activity which was further validated using colony forming assay and time killing kinetics. The duration of antifungal activity of peptide was improved using gelatin hydrogel through sustained delivery. RESULTS: VRF005 and VRF007 showed α-helical structure and was within the allowed region of Ramachandran plot. The simulation study showed their membrane penetration. The peptide uptake was found to be specific to corneal epithelial cells and also showed intracellular localization in Candida albicans and Fusarium solani. Peptides were found to be stable up to 2 hours when incubated with corneal epithelial tissue lysate. Dynamic light scattering, and native polyacrylamide gel electrophoresis revealed aggregation of peptides. VRF007 showed antifungal activity up to 24 hour whereas VRF005 showed activity up to 4 hours. Hence gelatin hydrogel-based delivery system was used to improve the activity. Actin staining of corneal epithelial cells showed that the cells were attached on gelatin hydrogel. CONCLUSION: We have designed corneal specific cell penetrating peptides using subtractive proteomic approach. Bioavailability and delivery of peptide was enhanced using gelatin hydrogel system.


Subject(s)
Antifungal Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Cornea/drug effects , Drug Delivery Systems , Drug Design , Gelatin/chemistry , Hydrogels/chemistry , Amino Acid Sequence , Antifungal Agents/chemistry , Candida albicans/cytology , Candida albicans/drug effects , Cell Adhesion/drug effects , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/toxicity , Colony Count, Microbial , Cornea/metabolism , Epithelial Cells/drug effects , Fusarium/drug effects , Humans , Kinetics , Molecular Dynamics Simulation
15.
Langmuir ; 35(9): 3513-3523, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30673275

ABSTRACT

In this work, we report a drug delivery system based on the pH-responsive self-assembly and -disassembly behaviors of peptides. Here, a systematically designed histidine-rich lipidated peptide (NP1) is presented to encapsulate and deliver an anticancer drug ellipticine (EPT) into two model cells: non-small-cell lung carcinoma and Chinese hamster ovary cells. The mechanism of pH-responsive peptide self-assembly and -disassembly involved in the drug encapsulation and release process are extensively investigated. We found that NP1 could self-assemble as a spherical nanocomplex (diameter = 34.43 nm) in a neutral pH environment with EPT encapsulated and positively charged arginine amino acids aligned outward and EPT is released in an acidic environment due to the pH-triggered disassembly. Furthermore, the EPT-encapsulating peptide could achieve a mass loading ability of 18% (mass of loaded-EPT/mass of NP1) with optimization. More importantly, it is revealed that the positively charged arginine on the periphery of the NP1 peptides could greatly facilitate their direct translocation through the negatively charged plasma membrane via electrostatic interaction, instead of via endocytosis, which provides a more efficient uptake pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/chemistry , Drug Carriers/chemistry , Ellipticines/pharmacology , Lipopeptides/chemistry , A549 Cells , Amino Acid Sequence , Animals , CHO Cells , Cell Survival/drug effects , Cell-Penetrating Peptides/toxicity , Cricetulus , Drug Carriers/toxicity , Drug Liberation , Histidine/chemistry , Humans , Hydrogen-Ion Concentration , Lipopeptides/toxicity , Nanostructures/chemistry , Nanostructures/toxicity
16.
J Am Chem Soc ; 140(50): 17484-17491, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30525541

ABSTRACT

Luminescent probes and nanoparticles (NPs) with long excited state lifetimes are essential for time-resolved biological imaging. Generally, cell membranes are physiological barriers that could prevent the uptake of many unnatural compounds. It is still a big challenge to prepare biocompatible imaging agents with high cytomembrane permeability, especially for nonmetallic NPs with long-lived luminescence. Herein, an amphiphilic cell-penetrating peptide, F6G6(rR)3R2, was designed to transport hydrophobic fluorophores across cellular barriers. Three classical thermally activated delayed fluorescence (TADF) molecules, 4CzIPN, NAI-DPAC, and BTZ-DMAC, could self-assemble into well-dispersed NPs with F6G6(rR)3R2 in aqueous solution. These NPs showed low cytotoxicity and could penetrate membranes easily. Moreover, long-lived TADF enabled them to be used in time-resolved luminescence imaging in oxygenic environments. These findings greatly expanded the applications of cell-penetrating peptides for delivery of molecules and NPs by only noncovalent interactions, which were more flexible and easier than covalent modifications.


Subject(s)
Cell-Penetrating Peptides/metabolism , Drug Carriers/metabolism , Fluorescent Dyes/chemistry , Nanoparticles/chemistry , 3T3 Cells , Animals , Cell Membrane/metabolism , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/toxicity , Drug Carriers/chemistry , Drug Carriers/toxicity , Fluorescence , Fluorescent Dyes/toxicity , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Nanoparticles/toxicity , Temperature
17.
J Am Chem Soc ; 140(49): 17234-17240, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30398334

ABSTRACT

The efficient delivery of proteins into cells is needed to fully realize the potential of protein-based therapeutics. Current protein delivery strategies generally suffer from poor endosomal escape and low tolerance for serum. Here, the genetic fusion of a supercharged polypeptide, called SCP, to a protein provides a generic method for intracellular protein delivery. It allows efficient protein endocytosis and endosomal escape and is capable of potently delivering various proteins with a range of charges, sizes, and bioactivities into the nucleus of living cells. SCP is discovered to bind directly to the nuclear import protein importin ß1 and gains access to the nucleus. Furthermore, SCP shows minimal hemolytic activity and stability in serum and lacks toxicity and immunogenicity in vivo. Effective gene editing can be achieved by SCP-mediated delivery of Cas9 protein and guide RNA. This study may provide an efficient and useful tool for the design and development of cell-nuclear-targeted drug delivery.


Subject(s)
Cell-Penetrating Peptides/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell-Penetrating Peptides/blood , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/toxicity , Endocytosis/physiology , Escherichia coli/genetics , Female , Humans , Mice, Inbred BALB C , Protein Stability , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/toxicity , beta Karyopherins/metabolism
18.
J Am Chem Soc ; 140(47): 16140-16151, 2018 11 28.
Article in English | MEDLINE | ID: mdl-30388366

ABSTRACT

New strategies are urgently needed to target MRSA, a major global health problem and the leading cause of mortality from antibiotic-resistant infections in many countries. Here, we report a general approach to this problem exemplified by the design and synthesis of a vancomycin-d-octaarginine conjugate (V-r8) and investigation of its efficacy in addressing antibiotic-insensitive bacterial populations. V-r8 eradicated MRSA biofilm and persister cells in vitro, outperforming vancomycin by orders of magnitude. It also eliminated 97% of biofilm-associated MRSA in a murine wound infection model and displayed no acute dermal toxicity. This new dual-function conjugate displays enhanced cellular accumulation and membrane perturbation as compared to vancomycin. Based on its rapid and potent activity against biofilm and persister cells, V-r8 is a promising agent against clinical MRSA infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Cell-Penetrating Peptides/therapeutic use , Methicillin-Resistant Staphylococcus aureus/physiology , Vancomycin/analogs & derivatives , Vancomycin/therapeutic use , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Cell Line , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/toxicity , Drug Design , Humans , Male , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice, Inbred C57BL , Microbial Sensitivity Tests , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Oligopeptides/toxicity , Vancomycin/pharmacology , Vancomycin/toxicity , Vancomycin-Resistant Enterococci/drug effects , Vancomycin-Resistant Enterococci/physiology
19.
20.
Sci Rep ; 8(1): 12538, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135446

ABSTRACT

Cell penetrating peptides (CPPs) offer great potential to deliver therapeutic molecules to previously inaccessible intracellular targets. However, many CPPs are inefficient and often leave their attached cargo stranded in the cell's endosome. We report a versatile platform for the isolation of peptides delivering a wide range of cargos into the cytoplasm of cells. We used this screening platform to identify multiple "Phylomer" CPPs, derived from bacterial and viral genomes. These peptides are amenable to conventional sequence optimization and engineering approaches for cell targeting and half-life extension. We demonstrate potent, functional delivery of protein, peptide, and nucleic acid analog cargos into cells using Phylomer CPPs. We validate in vivo activity in the cytoplasm, through successful transport of an oligonucleotide therapeutic fused to a Phylomer CPP in a disease model for Duchenne's muscular dystrophy. This report thus establishes a discovery platform for identifying novel, functional CPPs to expand the delivery landscape of druggable intracellular targets for biological therapeutics.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Drug Delivery Systems/methods , Drug Evaluation, Preclinical/methods , Animals , Bacteriophage T7 , Biotinylation , CHO Cells , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/toxicity , Circular Dichroism , Cricetulus , Disease Models, Animal , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Microscopy, Fluorescence , Muscular Dystrophy, Duchenne/drug therapy , Peptide Library , Repressor Proteins/genetics , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...