Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
Add more filters

Publication year range
1.
BMC Biol ; 22(1): 86, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38637801

ABSTRACT

BACKGROUND: The blood-brain barrier serves as a critical interface between the bloodstream and brain tissue, mainly composed of pericytes, neurons, endothelial cells, and tightly connected basal membranes. It plays a pivotal role in safeguarding brain from harmful substances, thus protecting the integrity of the nervous system and preserving overall brain homeostasis. However, this remarkable selective transmission also poses a formidable challenge in the realm of central nervous system diseases treatment, hindering the delivery of large-molecule drugs into the brain. In response to this challenge, many researchers have devoted themselves to developing drug delivery systems capable of breaching the blood-brain barrier. Among these, blood-brain barrier penetrating peptides have emerged as promising candidates. These peptides had the advantages of high biosafety, ease of synthesis, and exceptional penetration efficiency, making them an effective drug delivery solution. While previous studies have developed a few prediction models for blood-brain barrier penetrating peptides, their performance has often been hampered by issue of limited positive data. RESULTS: In this study, we present Augur, a novel prediction model using borderline-SMOTE-based data augmentation and machine learning. we extract highly interpretable physicochemical properties of blood-brain barrier penetrating peptides while solving the issues of small sample size and imbalance of positive and negative samples. Experimental results demonstrate the superior prediction performance of Augur with an AUC value of 0.932 on the training set and 0.931 on the independent test set. CONCLUSIONS: This newly developed Augur model demonstrates superior performance in predicting blood-brain barrier penetrating peptides, offering valuable insights for drug development targeting neurological disorders. This breakthrough may enhance the efficiency of peptide-based drug discovery and pave the way for innovative treatment strategies for central nervous system diseases.


Subject(s)
Cell-Penetrating Peptides , Central Nervous System Diseases , Humans , Blood-Brain Barrier/chemistry , Endothelial Cells , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/therapeutic use , Brain , Central Nervous System Diseases/drug therapy
2.
Molecules ; 29(6)2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38542884

ABSTRACT

Cell-penetrating peptides (CPPs) are invaluable tools for delivering various substances into cells by crossing biological membranes. However, the effects of cell-penetrating peptide fusion proteins on the biological activity of antibodies remain to be fully understood. Here, we engineered a recombinant protein, LP-scFv, which combines the single-chain variable region of anti-human epidermal growth factor receptor-2 with a novel and non-oxic cell-penetrating peptide as a leader peptide. The introduction of this leader peptide led to a more than twofold increase in the internalization efficiency of the single-chain antibody, as confirmed using microscopic analysis and flow cytometry. The effects of the single-chain antibodies and LP-scFv on cell viability were evaluated using the MTT assay. Both the single-chain antibodies and LP-scFv reduced the viability of BT474 and NCI-N87 cells in a dose-dependent manner while exhibiting minimal toxicity towards MCF-7 and MCF-10A cells. Further investigation into LP-scFv's mechanism revealed that the induced leader peptide does not alter the MAPK-ERK1/2 and PI3K/AKT pathways of single-chain antibodies. An enhanced antitumor activity was also confirmed in an NCI-N87 tumor xenograft model in mice with a reduction of 45.2% in tumor growth inhibition (vs. 23.1% for scFv) with a 50 mg/kg dose after orthotopic injection administration, which was equivalent to that of trastuzumab (vs. 55.7% for trastuzumab). Overall, these results indicate that LP-scFv exhibits significant permeation activity in HER2-positive cells to enhance the intracellular dose effect on antitumor activity in vitro and in vivo. This research lays the foundation for designing novel antibody-based therapies for cancer.


Subject(s)
Breast Neoplasms , Cell-Penetrating Peptides , Single-Chain Antibodies , Humans , Animals , Mice , Female , Breast Neoplasms/pathology , Single-Chain Antibodies/pharmacology , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Cell Line, Tumor , Trastuzumab/therapeutic use , Receptor, ErbB-2/metabolism , Protein Sorting Signals , Xenograft Model Antitumor Assays
3.
Proc Natl Acad Sci U S A ; 117(11): 6121-6128, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123072

ABSTRACT

Virus replication requires critical interactions between viral proteins and cellular proteins that mediate many aspects of infection, including the transport of viral genomes to the site of replication. In human papillomavirus (HPV) infection, the cellular protein complex known as retromer binds to the L2 capsid protein and sorts incoming virions into the retrograde transport pathway for trafficking to the nucleus. Here, we show that short synthetic peptides containing the HPV16 L2 retromer-binding site and a cell-penetrating sequence enter cells, sequester retromer from the incoming HPV pseudovirus, and inhibit HPV exit from the endosome, resulting in loss of viral components from cells and in a profound, dose-dependent block to infection. The peptide also inhibits cervicovaginal HPV16 pseudovirus infection in a mouse model. These results confirm the retromer-mediated model of retrograde HPV entry and validate intracellular virus trafficking as an antiviral target. More generally, inhibiting virus replication with agents that can enter cells and disrupt essential protein-protein interactions may be applicable in broad outline to many viruses.


Subject(s)
Capsid Proteins/metabolism , Cell-Penetrating Peptides/pharmacology , Human papillomavirus 16/drug effects , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/drug therapy , Virus Internalization/drug effects , Animals , Cell-Penetrating Peptides/therapeutic use , Cervix Uteri/virology , Disease Models, Animal , Female , HEK293 Cells , HeLa Cells , Human papillomavirus 16/physiology , Humans , Mice , Papillomavirus Infections/virology , Protein Binding/drug effects , Protein Interaction Maps/drug effects , Vagina/virology
4.
J Labelled Comp Radiopharm ; 66(9): 237-248, 2023 07.
Article in English | MEDLINE | ID: mdl-37002811

ABSTRACT

Trans-blood-brain barrier (BBB) delivery of therapeutic and diagnostic agents is a major challenge in the development of central nervous system (CNS) targeted radiopharmaceuticals. This review is an introduction to the use of peptides as delivery agents to transport cargos into the CNS. The most widely used BBB-penetrating peptides are reviewed here, with a particular emphasis on the broad range of cargos delivered into the CNS using these. Cell-penetrating peptides (CPPs) have been deployed as trans-BBB delivery agents for some time; new developments in the CPP field offer exciting opportunities for the design of next generation trans-BBB complexes. Many of the peptides highlighted here are ready to be combined with diagnostic and therapeutic radiopharmaceuticals to develop highly effective CNS-targeted agents.


Subject(s)
Blood-Brain Barrier , Cell-Penetrating Peptides , Radiopharmaceuticals , Drug Delivery Systems , Biological Transport , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/therapeutic use
5.
Int J Mol Sci ; 24(5)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36901707

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease (ND) and the leading cause of dementia. It is characterized by non-linear, genetic-driven pathophysiological dynamics with high heterogeneity in the biological alterations and the causes of the disease. One of the hallmarks of the AD is the progression of plaques of aggregated amyloid-ß (Aß) or neurofibrillary tangles of Tau. Currently there is no efficient treatment for the AD. Nevertheless, several breakthroughs in revealing the mechanisms behind progression of the AD have led to the discovery of possible therapeutic targets. Some of these include the reduction in inflammation in the brain, and, although highly debated, limiting of the aggregation of the Aß. In this work we show that similarly to the Neural cell adhesion molecule 1 (NCAM1) signal sequence, other Aß interacting protein sequences, especially derived from Transthyretin, can be used successfully to reduce or target the amyloid aggregation/aggregates in vitro. The modified signal peptides with cell-penetrating properties reduce the Aß aggregation and are predicted to have anti-inflammatory properties. Furthermore, we show that by expressing the Aß-EGFP fusion protein, we can efficiently assess the potential for reduction in aggregation, and the CPP properties of peptides in mammalian cells.


Subject(s)
Alzheimer Disease , Cell-Penetrating Peptides , Neurodegenerative Diseases , Animals , Humans , Cell-Penetrating Peptides/therapeutic use , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Protein Sorting Signals , tau Proteins/metabolism , Mammals/metabolism
6.
Int J Mol Sci ; 24(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36614268

ABSTRACT

Drug conjugates have become a significant focus of research in the field of targeted medicine for cancer treatments. Peptide-drug conjugates (PDCs), a subset of drug conjugates, are composed of carrier peptides ranging from 5 to 30 amino acid residues, toxic payloads, and linkers that connect the payload to the peptide. PDCs are further broken down into cell-penetrating peptides (CPPs) and cell-targeting peptides (CTPs), each having their own differences in the delivery of cytotoxic payloads. Generally, PDCs as compared to other drug conjugates-like antibody-drug conjugates (ADCs)-have advantages in tumor penetration, ease of synthesis and cost, and reduced off-target effects. Further, as compared to traditional cancer treatments (e.g., chemotherapy and radiation), PDCs have higher specificity for the target cancer with generally less toxic side effects in smaller doses. However, PDCs can have disadvantages such as poor stability and rapid renal clearance due to their smaller size and limited oral bioavailability due to digestion of its peptide structure. Some of these challenges can be overcome with modifications, and despite drawbacks, the intrinsic small size of PDCs with high target specificity still makes them an attractive area of research for cancer treatments.


Subject(s)
Antineoplastic Agents , Cell-Penetrating Peptides , Immunoconjugates , Neoplasms , Humans , Pharmaceutical Preparations/metabolism , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Neoplasms/metabolism , Immunoconjugates/therapeutic use , Cell-Penetrating Peptides/therapeutic use , Antigens/therapeutic use
8.
Mol Biol Rep ; 49(4): 3197-3212, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35094208

ABSTRACT

BACKGROUND: The antitumor peptide CIGB-552 is a new targeted anticancer therapy which molecular mechanism is associated with the inhibition of the transcription factor NF-kB, mediated by COMMD1 protein stabilization. In this study, we examined the antiproliferative capacity of CIGB-552 in combination with chemotherapeutic agents in lung cancer models. METHODS AND RESULTS: We combined of CIGB-552 and the antineoplastic agent Cisplatin (CDDP) in concomitant and pre-treatment scenary in a dose matrix approach. This study was performed in the non-small cell lung cancer cell lines NCI-H460, A549 and in a mouse model of TC-1 lung cancer. Our results demonstrate a clear synergic effect between 37.5 µM of CIGB-552 and 5 µM of CDDP under concomitant scheme, on proliferation inhibition, cell cycle arrest, apoptosis induction and oxidative stress response. The effect of CIGB-552 (1 mg/kg) and CDDP (0.4 mg/kg) administrated as a combined therapy was demonstrated in vivo in a TC-1 mouse model where the combination achieved an effective antitumor response, without any deterioration signs or side effects. CONCLUSIONS: These findings demonstrate the efficacy of the concomitant combination of both drugs in preclinical studies and support the use of this therapy in clinical trials. This study is the first evidence of synergistic effect of the combination of  the antitumoral peptide CIGB-552 and CDDP.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/therapeutic use , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Synergism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Peptides/metabolism
9.
Cell Mol Biol Lett ; 27(1): 33, 2022 Apr 09.
Article in English | MEDLINE | ID: mdl-35397496

ABSTRACT

BACKGROUND: Nowadays, conventional medical treatments such as surgery, radiotherapy, and chemotherapy cannot cure all types of cancer. A promising approach to treat solid tumors is the use of tumor-targeting peptides to deliver drugs or active agents selectively. RESULT: Introducing beneficial therapeutic approaches, such as therapeutic peptides and their varied methods of action against tumor cells, can aid researchers in the discovery of novel peptides for cancer treatment. The biomedical applications of therapeutic peptides are highly interesting. These peptides, owing to their high selectivity, specificity, small dimensions, high biocompatibility, and easy modification, provide good opportunities for targeted drug delivery. In recent years, peptides have shown considerable promise as therapeutics or targeting ligands in cancer research and nanotechnology. CONCLUSION:  This study reviews a variety of therapeutic peptides and targeting ligands in cancer therapy. Initially, three types of tumor-homing and cell-penetrating peptides (CPPs) are described, and then their applications in breast, glioma, colorectal, and melanoma cancer research are discussed.


Subject(s)
Antineoplastic Agents , Cell-Penetrating Peptides , Glioma , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/therapeutic use , Drug Delivery Systems/methods , Glioma/drug therapy , Humans , Ligands , Neoplasms/drug therapy
10.
Mar Drugs ; 20(12)2022 Dec 04.
Article in English | MEDLINE | ID: mdl-36547910

ABSTRACT

Complex pathological diseases, such as cancer, infection, and Alzheimer's, need to be targeted by multipronged curative. Various omics technologies, with a high rate of data generation, demand artificial intelligence to translate these data into druggable targets. In this study, 82 marine venomous animal species were retrieved, and 3505 cryptic cell-penetrating peptides (CPPs) were identified in their toxins. A total of 279 safe peptides were further analyzed for antimicrobial, anticancer, and immunomodulatory characteristics. Protease-resistant CPPs with endosomal-escape ability in Hydrophis hardwickii, nuclear-localizing peptides in Scorpaena plumieri, and mitochondrial-targeting peptides from Synanceia horrida were suitable for compartmental drug delivery. A broad-spectrum S. horrida-derived antimicrobial peptide with a high binding-affinity to bacterial membranes was an antigen-presenting cell (APC) stimulator that primes cytokine release and naïve T-cell maturation simultaneously. While antibiofilm and wound-healing peptides were detected in Synanceia verrucosa, APC epitopes as universal adjuvants for antiviral vaccination were in Pterois volitans and Conus monile. Conus pennaceus-derived anticancer peptides showed antiangiogenic and IL-2-inducing properties with moderate BBB-permeation and were defined to be a tumor-homing peptide (THP) with the ability to inhibit programmed death ligand-1 (PDL-1). Isoforms of RGD-containing peptides with innate antiangiogenic characteristics were in Conus tessulatus for tumor targeting. Inhibitors of neuropilin-1 in C. pennaceus are proposed for imaging probes or therapeutic delivery. A Conus betulinus cryptic peptide, with BBB-permeation, mitochondrial-targeting, and antioxidant capacity, was a stimulator of anti-inflammatory cytokines and non-inducer of proinflammation proposed for Alzheimer's. Conclusively, we have considered the dynamic interaction of cells, their microenvironment, and proportional-orchestrating-host- immune pathways by multi-target-directed CPPs resembling single-molecule polypharmacology. This strategy might fill the therapeutic gap in complex resistant disorders and increase the candidates' clinical-translation chance.


Subject(s)
Alzheimer Disease , Anti-Infective Agents , Cell-Penetrating Peptides , Neoplasms , Animals , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/therapeutic use , Venoms , Artificial Intelligence , Polypharmacology , Anti-Infective Agents/pharmacology , Anti-Infective Agents/therapeutic use , Neoplasms/drug therapy , Tumor Microenvironment
11.
Cancer Sci ; 112(6): 2118-2125, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33793015

ABSTRACT

Cell-penetrating peptides, such as antibodies, have gained great attention as tools for the development of specific delivery systems for payloads, which might be applied as non-invasive carriers in vivo. Among these, tumor-homing peptides recently have been studied for use in tumor medicine. Tumor-homing peptides are oligopeptides, usually consisting of 30 or fewer amino acids that are efficiently and specifically incorporated into tumor cells, suggesting their potential use in establishing novel non-invasive tumor imaging systems for diagnostic and therapeutic applications. Here, we briefly introduce the biological characteristics of our tumor-homing peptides, focusing especially on those developed using a random peptide library constructed using mRNA display technology. The advantage of the tumor-homing peptides is their biological safety, given that these molecules do not show significant cytotoxicity against non-neoplastic cells; lack serious antigenicity, which alternatively might evoke unfavorable immune responses and inflammation in vivo; and are rapidly incorporated into target cells/tissues, with rates exceeding those seen for antibodies. Given their small size, tumor-homing peptides also are easy to modify and redesign. Based on these merits, tumor-homing peptides are expected to find wide application in various aspects of tumor medicine, including imaging diagnostics (eg, with dye-conjugated probes for direct visualization of invasive/metastatic tumor lesions in vivo) and therapeutics (eg, using peptide-drug conjugates [PDCs] for tumor targeting). Although further evidence will be required to demonstrate their practical utility, tumor-homing peptides are expected to show great potential as a next-generation bio-tool contributing to precision medicine for cancer patients.


Subject(s)
Cell-Penetrating Peptides/physiology , Cell-Penetrating Peptides/therapeutic use , Neoplasms/diagnosis , Neoplasms/therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell-Penetrating Peptides/chemistry , Drug Delivery Systems , Humans , Neoplasms/metabolism , Oligopeptides/chemistry , Oligopeptides/physiology , Oligopeptides/therapeutic use , Peptide Library , Precision Medicine , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Pharmacol Res ; 167: 105441, 2021 05.
Article in English | MEDLINE | ID: mdl-33503478

ABSTRACT

As described by Jean Martin Charcot in 1868, multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease of the central nervous system (CNS) which leads to permanent disability in patients. Following CNS insults, astrocytes and microglial cells undergo changes, which lead to scar formation in the site of injury. Owning to the pathophysiology of MS lesions, changes in both cellular and extracellular matrix (ECM) components occur over the progression of disease. In spite of advances in therapeutic approaches, drug delivery to MS lesions appears of great interest with big challenges and limitations. Targeting with peptides is a novel promising approach in the field of drug delivery. Recently peptides have been used for active targeting of different pathological disorders in which specific peptides make targeted accumulation of cargos to enhance local drug concentration at the pathological area, lead to increased therapeutic efficacy and decreased side effects. However, specific approaches for targeting the lesion in MS are still lacking. In this review, we discuss the changes of the ECM components as well as the cellular characteristics of demyelinated lesions and emphasis on opportunities for peptide based targeted drug delivery to highlight the possibility of such approaches for neurodegenerative disease with specific focus on MS.


Subject(s)
Brain/drug effects , Cell-Penetrating Peptides/metabolism , Drug Carriers/metabolism , Drug Delivery Systems/methods , Multiple Sclerosis/drug therapy , Animals , Brain/metabolism , Brain/pathology , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/therapeutic use , Drug Carriers/chemistry , Drug Carriers/therapeutic use , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology
13.
J Nanobiotechnology ; 19(1): 255, 2021 Aug 23.
Article in English | MEDLINE | ID: mdl-34425832

ABSTRACT

Although nanomedicine have greatly developed and human life span has been extended, we have witnessed the soared incidence of central nervous system (CNS) diseases including neurodegenerative diseases (Alzheimer's disease, Parkinson's disease), ischemic stroke, and brain tumors, which have severely damaged the quality of life and greatly increased the economic and social burdens. Moreover, partial small molecule drugs and almost all large molecule drugs (such as recombinant protein, therapeutic antibody, and nucleic acid) cannot cross the blood-brain barrier. Therefore, it is especially important to develop a drug delivery system that can effectively deliver therapeutic drugs to the central nervous system for the treatment of central nervous system diseases. Cell penetrating peptides (CPPs) provide a potential strategy for the transport of macromolecules through the blood-brain barrier. This study analyzed and summarized the progress of CPPs in CNS diseases from three aspects: CPPs, the conjugates of CPPs and drug, and CPPs modified nanoparticles to provide scientific basis for the application of CPPs for CNS diseases.


Subject(s)
Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/therapeutic use , Central Nervous System Diseases/drug therapy , Nanostructures/chemistry , Nanostructures/therapeutic use , Alzheimer Disease , Animals , Blood-Brain Barrier/metabolism , Brain Neoplasms , Drug Delivery Systems , Glioma , Humans , Ischemic Stroke , Nanomedicine , Nanoparticles/chemistry , Parkinson Disease , Quality of Life , Recombinant Proteins
14.
Int J Mol Sci ; 22(22)2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34830481

ABSTRACT

Therapies for stroke have remained elusive in the past despite the great relevance of this pathology. However, recent results have provided strong evidence that postsynaptic density protein-95 (PSD-95) can be exploited as an efficient target for stroke neuroprotection by strategies able to counteract excitotoxicity, a major mechanism of neuronal death after ischemic stroke. This scaffold protein is key to the maintenance of a complex framework of protein interactions established at the postsynaptic density (PSD) of excitatory neurons, relevant to neuronal function and survival. Using cell penetrating peptides (CPPs) as therapeutic tools, two different approaches have been devised and advanced to different levels of clinical development. First, nerinetide (Phase 3) and AVLX-144 (Phase 1) were designed to interfere with the coupling of the ternary complex formed by PSD-95 with GluN2B subunits of the N-methyl-D-aspartate type of glutamate receptors (NMDARs) and neuronal nitric oxide synthase (nNOS). These peptides reduced neurotoxicity derived from NMDAR overactivation, decreased infarct volume and improved neurobehavioral results in different models of ischemic stroke. However, an important caveat to this approach was PSD-95 processing by calpain, a pathological mechanism specifically induced by excitotoxicity that results in a profound alteration of survival signaling. Thus, a third peptide (TP95414) has been recently developed to interfere with PSD-95 cleavage and reduce neuronal death, which also improves neurological outcome in a preclinical mouse model of permanent ischemia. Here, we review recent advancements in the development and characterization of PSD-95-targeted CPPs and propose the combination of these two approaches to improve treatment of stroke and other excitotoxicity-associated disorders.


Subject(s)
Cell-Penetrating Peptides/therapeutic use , Disks Large Homolog 4 Protein/genetics , Ischemic Stroke/drug therapy , Neuroprotective Agents/therapeutic use , Calpain/genetics , Cell-Penetrating Peptides/genetics , Disks Large Homolog 4 Protein/antagonists & inhibitors , Humans , Ischemic Stroke/genetics , Ischemic Stroke/pathology , Protein Interaction Maps/genetics
15.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946854

ABSTRACT

Hexokinases are a family of ubiquitous exose-phosphorylating enzymes that prime glucose for intracellular utilization. Hexokinase 2 (HK2) is the most active isozyme of the family, mainly expressed in insulin-sensitive tissues. HK2 induction in most neoplastic cells contributes to their metabolic rewiring towards aerobic glycolysis, and its genetic ablation inhibits malignant growth in mouse models. HK2 can dock to mitochondria, where it performs additional functions in autophagy regulation and cell death inhibition that are independent of its enzymatic activity. The recent definition of HK2 localization to contact points between mitochondria and endoplasmic reticulum called Mitochondria Associated Membranes (MAMs) has unveiled a novel HK2 role in regulating intracellular Ca2+ fluxes. Here, we propose that HK2 localization in MAMs of tumor cells is key in sustaining neoplastic progression, as it acts as an intersection node between metabolic and survival pathways. Disrupting these functions by targeting HK2 subcellular localization can constitute a promising anti-tumor strategy.


Subject(s)
Hexokinase/physiology , Neoplasm Proteins/physiology , Neoplasms/enzymology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Autophagy/physiology , Calcium Signaling/physiology , Cell Hypoxia , Cell-Penetrating Peptides/therapeutic use , Enzyme Induction , Gene Expression Regulation, Neoplastic , Glycolysis/physiology , Hexokinase/antagonists & inhibitors , Humans , Intracellular Membranes/enzymology , Mice , MicroRNAs/genetics , Mitochondria/metabolism , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/therapy , Neoplasms, Experimental/enzymology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational , Rats , Ubiquitination
16.
Molecules ; 26(2)2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33445797

ABSTRACT

A major obstacle in tumor treatment is associated with the poor penetration of a therapeutic agent into the tumor tissue and with their adverse influence on healthy cells, which limits the dose of drug that can be safely administered to cancer patients. Gemcitabine is an anticancer drug used to treat a wide range of solid tumors and is a first-line treatment for pancreatic cancer. The effect of gemcitabine is significantly weakened by its rapid plasma degradation. In addition, the systemic toxicity and drug resistance significantly reduce its chemotherapeutic efficacy. Up to now, many approaches have been made to improve the therapeutic index of gemcitabine. One of the recently developed approaches to improve conventional chemotherapy is based on the direct targeting of chemotherapeutics to cancer cells using the drug-peptide conjugates. In this work, we summarize recently published gemcitabine peptide-based conjugates and their efficacy in anticancer therapy.


Subject(s)
Deoxycytidine/analogs & derivatives , Molecular Targeted Therapy , Neoplasms/drug therapy , Peptides/therapeutic use , Animals , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/therapeutic use , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Humans , Nanoparticles/chemistry , Peptides/pharmacology , Gemcitabine
17.
Pharmacol Res ; 155: 104750, 2020 05.
Article in English | MEDLINE | ID: mdl-32151680

ABSTRACT

Pepducins are cell-penetrating, membrane-tethered lipopeptides designed to target the intracellular region of a G protein-coupled receptor (GPCR) in order to allosterically modulate the receptor's signaling output. In this proof-of-concept study, we explored the pain-relief potential of a pepducin series derived from the first intracellular loop of neurotensin receptor type 1 (NTS1), a class A GPCR that mediates many of the effects of the neurotensin (NT) tridecapeptide, including hypothermia, hypotension and analgesia. We used BRET-based biosensors to determine the pepducins' ability to engage G protein signaling pathways associated with NTS1 activation. We observed partial Gαq and Gα13 activation at a 10 µM concentration, indicating that these pepducins may act as allosteric agonists of NTS1. Additionally, we used surface plasmon resonance (SPR) as a label-free assay to monitor pepducin-induced responses in CHO-K1 cells stably expressing hNTS1. This whole-cell integrated assay enabled us to subdivide our pepducin series into three profile response groups. In order to determine the pepducins' antinociceptive potential, we then screened the series in an acute pain model (tail-flick test) by measuring tail withdrawal latencies to a thermal nociceptive stimulus, following intrathecal (i.t.) pepducin administration (275 nmol/kg). We further evaluated promising pepducins in a tonic pain model (formalin test), as well as in neuropathic (Chronic Constriction Injury) and inflammatory (Complete Freund's Adjuvant) chronic pain models. We report one pepducin, PP-001, that consistently reduced rat nociceptive behaviors, even in chronic pain paradigms. Finally, we designed a TAMRA-tagged version of PP-001 and found by confocal microscopy that the pepducin reached the rat dorsal root ganglia post i.t. injection, thus potentially modulating the activity of NTS1 at this location to produce its analgesic effect. Altogether, these results suggest that NTS1-derived pepducins may represent a promising strategy in pain-relief.


Subject(s)
Analgesics/therapeutic use , Cell-Penetrating Peptides/therapeutic use , Lipopeptides/therapeutic use , Pain/drug therapy , Receptors, Neurotensin , Analgesics/pharmacology , Animals , CHO Cells , Cell-Penetrating Peptides/pharmacology , Cricetulus , GTP-Binding Proteins/metabolism , Ganglia, Spinal/metabolism , Lipopeptides/pharmacology , Male , Pain/genetics , Pain/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects
18.
Molecules ; 25(22)2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33187056

ABSTRACT

Amyloid proteins are linked to the pathogenesis of several diseases including Alzheimer's disease, but at the same time a range of functional amyloids are physiologically important in humans. Although the disease pathogenies have been associated with protein aggregation, the mechanisms and factors that lead to protein aggregation are not completely understood. Paradoxically, unique characteristics of amyloids provide new opportunities for engineering innovative materials with biomedical applications. In this review, we discuss not only outstanding advances in biomedical applications of amyloid peptides, but also the mechanism of amyloid aggregation, factors affecting the process, and core sequences driving the aggregation. We aim with this review to provide a useful manual for those who engineer amyloids for innovative medicine solutions.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/metabolism , Protein Aggregates , Alzheimer Disease/therapy , Amino Acid Motifs , Amyloidosis/metabolism , Amyloidosis/therapy , Animals , Antiviral Agents/pharmacology , Binding Sites , Cell-Penetrating Peptides/therapeutic use , Humans , Ions , Metals/chemistry , Nanomedicine , Protein Binding , Virus Diseases/drug therapy
19.
Arch Biochem Biophys ; 661: 74-86, 2019 01.
Article in English | MEDLINE | ID: mdl-30447207

ABSTRACT

In 1988, two unrelated papers reported the discovery of peptide vectors with innate cell translocation properties, setting the ground for a new area of research that over the years has grown into considerable therapeutic potential. The vectors, named cell-penetrating peptides (CPPs), constitute a now large and diversified family, sharing the extraordinary ability to diffuse unaltered across cell membranes while ferrying diverse associated cargos. Such properties have made CPPs ideal tools for delivery of nucleic acids, proteins and other therapeutic/diagnostic molecules to cells and tissues via covalent conjugation or complexation. This year 2018 marks the 30th anniversary of a peptide research landmark opening new perspectives in drug delivery. Given its vastness, exhaustive coverage of the main features and accomplishments in the CPP field is virtually impossible. Hence this manuscript, after saluting the above 30th jubilee, focuses by necessity on the most recent contributions, providing a comprehensive list of recognized CPPs and their latest-reported applications over the last two years. In addition, it thoroughly reviews three areas of peptide vector research of particular interest to us, namely (i) efficient transport of low-bioavailability drugs into the brain; (ii) CPP-delivered disruptors of G protein-coupled receptor (GPCRs) heteromers related to several disorders, and (iii) CPP-mediated delivery of useful but poorly internalized drugs into parasites.


Subject(s)
Biomedical Research/history , Cell-Penetrating Peptides/history , Cell-Penetrating Peptides/therapeutic use , Drug Delivery Systems/history , Drug Delivery Systems/methods , Animals , History, 20th Century , History, 21st Century , Humans
20.
Mol Pharm ; 16(3): 1140-1155, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30668131

ABSTRACT

Chronic inflammation is closely related to the development, deterioration, and metastasis of tumors. Recently, many studies have shown that down-regulating the expression of inflammation by blocking nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways could significantly inhibit tumor growth and metastasis. The combined application of curcumin (CUR) and celecoxib (CXB) has been proven to exert a synergistic antitumor effect via inhibiting the activation of NF-κB and STAT3. TAT-NBD (TN) peptide, a fusion peptide of NF-κB essential modulator (NEMO)-binding domain peptide (NBD) and cell-penetrating peptide (TAT), can selectively block NF-κB activating pathway resulting in tumor growth inhibition. In the present study, a novel TN-modified liposome coloading both CXB and CUR (TN-CCLP) at a synergistic ratio was first constructed with the property of synchronous release, then hyaluronic acid (HA) as CD44 targeting moiety was coated on the surface of the cationic liposome via electrostatic interaction to prepare the anionic HA/TN-CCLP. In vitro results of cytotoxicity, macrophage migration inhibition, and anti-inflammation efficacy revealed that TN-CCLP and HA/TN-CCLP were significantly superior to TN-LP and CCLP, while TN-CCLP exhibited better effects than HA/TN-CCLP due to higher cellular uptake ability. Different from in vitro data, after systematically treating 4T1 breast tumor-bearing mice, HA/TN-CCLP exerted the most striking effects on anti-inflammation, inhibition of macrophage recruitment, and antitumor because of the longest circulation time and maximum tumor accumulation. In particular, HA/TN-CCLP could availably block the lung metastasis of breast cancer. Taken together, the novel CD44 targeted TN-CCLP exhibited the potential for inhibiting tumor development and metastasis through improving inflammatory infiltration of tumor tissue.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/therapeutic use , Drug Therapy, Combination/methods , Hyaluronic Acid/chemistry , Inflammatory Breast Neoplasms/drug therapy , NF-kappa B/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Celecoxib/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Curcumin/therapeutic use , Drug Liberation , Drug Synergism , Female , Heterografts , Humans , Hyaluronan Receptors/metabolism , Inflammatory Breast Neoplasms/pathology , Liposomes , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Treatment Outcome , Tumor Burden
SELECTION OF CITATIONS
SEARCH DETAIL