Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Type of study
Publication year range
1.
Biomed Pharmacother ; 174: 116573, 2024 May.
Article in English | MEDLINE | ID: mdl-38613996

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by the absence of commonly targeted receptors. Unspecific chemotherapy is currently the main therapeutic option, with poor results. Another major challenge is the frequent appearance of brain metastasis (BM) associated with a significant decrease in patient overall survival. The treatment of BM is even more challenging due to the presence of the blood-brain barrier (BBB). Here, we present a dual-acting peptide (PepH3-vCPP2319) designed to tackle TNBC/BM, in which a TNBC-specific anticancer peptide (ACP) motif (vCPP2319) is joined to a BBB peptide shuttle (BBBpS) motif (PepH3). PepH3-vCPP2319 demonstrated selectivity and efficiency in eliminating TNBC both in monolayers (IC50≈5.0 µM) and in spheroids (IC50≈25.0 µM), with no stringent toxicity toward noncancerous cell lines and red blood cells (RBCs). PepH3-vCPP2319 was also able to cross the BBB in vitro and penetrate the brain in vivo, and was stable in serum with a half-life above 120 min. Tumor cell-peptide interaction is fast, with quick peptide internalization via clathrin-mediated endocytosis without membrane disruption. Upon internalization, the peptide is detected in the nucleus and the cytoplasm, indicating a multi-targeted mechanism of action that ultimately induces irreversible cell damage and apoptosis. In conclusion, we have designed a dual-acting peptide capable of brain penetration and TNBC cell elimination, thus expanding the drug arsenal to fight this BC subtype and its BM.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Peptides , Triple Negative Breast Neoplasms , Humans , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Brain Neoplasms/pathology , Female , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Cell Line, Tumor , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Animals , Peptides/pharmacology , Antineoplastic Agents/pharmacology , Endocytosis/drug effects
2.
J Thromb Haemost ; 22(4): 1046-1055, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38159648

ABSTRACT

BACKGROUND: COVID-19 vaccines have been widely used to control the SARS-CoV-2 pandemic. In individuals receiving replication-incompetent, adenovirus vector-based COVID-19 vaccines (eg, ChAdOx1 nCoV-19 [AstraZeneca] or Ad26.COV2.S [Johnson & Johnson/Janssen] vaccines), a very rare but serious adverse reaction has been reported and described as vaccine-induced immune thrombotic thrombocytopenia (VITT). The exact mechanism of VITT following Ad26.COV2.S vaccination is under investigation. Antibodies directed against human platelet factor 4 (PF4) are considered critical in the pathogenesis of VITT, suggesting similarities with heparin-induced thrombocytopenia. It has been postulated that components of these vaccines mimic the role of heparin by binding to PF4, triggering production of these anti-PF4 antibodies. OBJECTIVES: This study aimed to investigate the potential interaction between human PF4 and Ad26.COV2.S vaccine using several biophysical techniques. METHODS: Direct interaction of PF4 with Ad26.COV2.S vaccine was investigated using dynamic light scattering, biolayer interferometry, and surface plasmon resonance. For both biosensing methods, the Ad26.COV2.S vaccine was immobilized to the sensor surface and PF4 was used as analyte. RESULTS: No direct interactions between PF4 and Ad26.COV2.S vaccine could be detected using dynamic light scattering and biolayer interferometry. Surface plasmon resonance technology was shown to be unsuitable to investigate these types of interactions. CONCLUSION: Our findings make it very unlikely that direct binding of PF4 to Ad26.COV2.S vaccine or components thereof is driving the onset of VITT, although the occurrence of such interactions after immunization (potentially facilitated by unknown plasma or cellular factors) cannot be excluded. Further research is warranted to improve the understanding of the full mechanism of this adverse reaction.


Subject(s)
COVID-19 , Purpura, Thrombocytopenic, Idiopathic , Thrombocytopenia , Vaccines , Humans , Ad26COVS1 , Platelet Factor 4 , COVID-19 Vaccines/adverse effects , ChAdOx1 nCoV-19 , COVID-19/prevention & control , SARS-CoV-2 , Immunologic Factors
3.
Biomedicines ; 10(9)2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36140211

ABSTRACT

Ctn[15-34], the C-terminal section of crotalicidin (Ctn), a cathelicidin from a South American pit viper, is an antimicrobial and antitumoral peptide with remarkably longer stability in human serum than the parent Ctn. In this work, a set of topoisomers of both Ctn and Ctn[15-34], including the retro, enantio, and retroenantio versions, were synthesized and tested to investigate the structural requirements for activity. All topoisomers were as active as the cognate sequences against Gram-negative bacteria and tumor cells while slightly more toxic towards normal cells. More importantly, the enhanced serum stability of the D-amino-acid-containing versions suggests that such topoisomers must be preferentially considered as future antimicrobial and anticancer peptide leads.

4.
ACS Infect Dis ; 7(1): 6-22, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33319557

ABSTRACT

There is an urgent need for the development of new anti-HIV drugs that can complement existing medicines to be used against resistant strains. Here, we report the anti-HIV-1 peptide pepRF1, a human serum-resistant peptide derived from the Dengue virus capsid protein. In vitro, pepRF1 shows a 50% inhibitory concentration of 1.5 nM with a potential therapeutic window higher than 53 000. This peptide is specific for CXCR4-tropic strains, preventing viral entry into target cells by binding to the viral coreceptor CXCR4, acting as an antagonist of this receptor. pepRF1 is more effective than T20, the only peptide-based HIV-1 entry inhibitor approved, and excels in inhibiting a HIV-1 strain resistant to T20. Potentially, pepRF1 can be used alone or in combination with other anti-HIV drugs. Furthermore, one can also envisage its use as a novel therapeutic strategy for other CXCR4-related diseases.


Subject(s)
Dengue Virus , HIV Infections , HIV-1 , Capsid Proteins/genetics , Humans , Proteolysis , Receptors, CXCR4
5.
Front Bioeng Biotechnol ; 8: 552035, 2020.
Article in English | MEDLINE | ID: mdl-33015016

ABSTRACT

The characterization of biologically active peptides relies heavily on the study of their efficacy, toxicity, mechanism of action, cellular uptake, or intracellular location, using both in vitro and in vivo studies. These studies frequently depend on the use of fluorescence-based techniques. Since most peptides are not intrinsically fluorescent, they are conjugated to a fluorophore. The conjugation may interfere with peptide properties, thus biasing the results. The selection of the most suitable fluorophore is highly relevant. Here, a comprehensive study with blood-brain barrier (BBB) peptide shuttles (PepH3 and PepNeg) and antimicrobial peptides (AMPs) (vCPP2319 and Ctn[15-34]), tested as anticancer peptides (ACPs), having different fluorophores, namely 5(6)-carboxyfluorescein (CF), rhodamine B (RhB), quasar 570 (Q570), or tide fluor 3 (TF3) attached is presented. The goal is the evaluation of the impact of the selected fluorophores on peptide performance, applying routinely used techniques to assess cytotoxicity/toxicity, secondary structure, BBB translocation, and cellular internalization. Our results show that some fluorophores significantly modulate peptide activity when compared with unlabeled peptides, being more noticeable in hydrophobic and charged fluorophores. This study highlights the need for a careful experimental design for fluorescently labeled molecules, such as peptides.

6.
ACS Chem Biol ; 15(11): 2945-2957, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33021779

ABSTRACT

Crotalicidin (Ctn) and its fragment Ctn[15-34] are snake-venom-derived, cathelicidin-related peptides outstanding for their promising antimicrobial, antifungal, and antitumoral properties. In this study, we describe their membranolytic mechanisms as well as their putative interference with intracellular targets, both contributing to their antitumoral action against a pro-monocytic leukemia cell line. Initial flow cytometry assays demonstrated peptide ability to induce tumor cell membrane permeabilization and caspase-dependent apoptosis, without total activity reduction by serum proteases up to 24 h (Ctn) and 18 h (Ctn[15-34]). In addition, both Ctn and Ctn[15-34] showed preference for tumor cells rather than healthy cells, with selectivity ratios (tumoral vs healthy cells) of 17 and 7, respectively. Further microscopy and flow cytometry studies suggested their preferential accumulation in the cytoplasmic membrane and nucleus and proposed multiple predominant routes of peptide uptake, including direct entry and endocytosis. Affinity purification followed by proteomic identification experiments revealed both peptides to interact with proteins involved in DNA and protein metabolism, cell cycles, signal transduction, and/or programmed cell death, among others. These results suggest a putative role of Ctn and Ctn[15-34] to interact with key intracellular pathways, ultimately contributing to tumor cell death by necrosis/apoptosis. Altogether, this work proposes a dual mechanism underlying the antitumoral activity of Ctn and Ctn[15-34] and reinforces their potential as future therapeutic drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane Permeability/drug effects , Neoplasms/drug therapy , Peptide Fragments/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/pathology , Humans , Neoplasms/pathology , Peptide Fragments/chemistry
7.
Toxins (Basel) ; 12(4)2020 04 15.
Article in English | MEDLINE | ID: mdl-32326531

ABSTRACT

For decades, natural products in general and snake venoms (SV) in particular have been a rich source of bioactive compounds for drug discovery, and they remain a promising substrate for therapeutic development. Currently, a handful of SV-based drugs for diagnosis and treatment of various cardiovascular disorders and blood abnormalities are on the market. Likewise, far more SV compounds and their mimetics are under investigation today for diverse therapeutic applications, including antibiotic-resistant bacteria and cancer. In this review, we analyze the state of the art regarding SV-derived compounds with therapeutic potential, focusing on the development of antimicrobial and anticancer drugs. Specifically, information about SV peptides experimentally validated or predicted to act as antimicrobial and anticancer peptides (AMPs and ACPs, respectively) has been collected and analyzed. Their principal activities both in vitro and in vivo, structures, mechanisms of action, and attempts at sequence optimization are discussed in order to highlight their potential as drug leads.


Subject(s)
Anti-Bacterial Agents , Antineoplastic Agents , Peptides , Snake Venoms , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Humans , Neoplasms/drug therapy , Peptides/administration & dosage , Peptides/chemistry , Snake Venoms/administration & dosage , Snake Venoms/chemistry
8.
J Pept Sci ; 25(8): e3195, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31317613

ABSTRACT

Ctn[15-34], a downsized version of the snake venom cathelicidin-like peptide crotalicidin (Ctn), shows an unusually high lifespan (t1/2 , approximately 12 h) in human serum, which significantly adds to its promise as an antimicrobial and antitumor agent. Herein we investigate the role of Ctn[15-34] structure on serum survival. Using a set of analogs, we show that C-terminal amidation, as well as the specific layout of the Ctn[15-34] sequence-a helical N-terminal domain followed by a hydrophobic domain-is crucial for slow degradation, and any change in their arrangement results in significantly lower t1/2 . Aside from the privileged primary structure, features such as self-aggregation can be ruled out as causes for the long serum life. Instead, studies in other protease-rich fluids suggest a key role for certain human serum components. Finally, we demonstrate that Ctn[15-34] is able to induce bacterial death even after 12-hour pre-incubation in serum, in agreement with the proteolytic data. Altogether, the results shed light on the uncommon stability of Ctn[15-34] in human serum and confirm its potential as an anti-infective lead.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Escherichia coli/drug effects , Peptide Fragments/pharmacology , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/blood , Antimicrobial Cationic Peptides/chemistry , Escherichia coli/growth & development , Humans , Microbial Sensitivity Tests , Peptide Fragments/blood , Peptide Fragments/chemistry , Protein Conformation , Protein Stability
9.
J Proteomics ; 204: 103372, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31051282

ABSTRACT

The inherent propensity to enzymatic degradation of most peptides remains a bottleneck in their therapeutic development. Efficient, early screening methods are necessary for in vitro characterization of the molecular events occurring when peptides get in contact with biological fluids such us plasma. Herein we present an affinity purification/MS approach for mapping peptide serum interactors. We have applied this methodology to identify the serum partners of antibiotic peptide Ctn [15-34], aiming to ascertain the molecular interactions underlying its unusually long half-life (~ 12 h) in human serum. From 42 proteins captured in pull-downs with biotinylated Ctn [15-34] as bait, five are of special interest for their transport/binding properties hence alleged peptide arresting potential. The subset contains two members of the albumin superfamily, two apolipoproteins and a globulin. All five share a binding ability for hydrophobic species, and also bind Ctn [15-34], presumably via its C-terminal hydrophobic section, with affinities in the µM range as shown by surface plasmon resonance. Additionally, our functional enrichment reveals several significant immune-related processes suggesting an immunomodulatory role of Ctn [15-34]. Taken together, this study exemplifies how pharmacoproteomics can be used to analyze bioavailability issues and shed light on the serum interactors ultimately conferring protection to Ctn [15-34] against proteolytic events. SIGNIFICANCE: The affinity purification/MS identification methodology reported here can be viewed as a routine pharmacoproteomic approach to investigate the serum interactome of peptide drugs, identifying proteins affecting bioavailability and thus assisting the peptide drug development process. The specific results described here enlighten the serum stability issues of peptide Ctn [15-34] and ratify its promising future as an anti-infective lead.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Blood Proteins/chemistry , Reptilian Proteins/chemistry , Snake Venoms/chemistry , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Half-Life , Humans , Reptilian Proteins/metabolism
10.
J Biol Chem ; 293(5): 1536-1549, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29255091

ABSTRACT

Crotalicidin (Ctn), a cathelicidin-related peptide from the venom of a South American rattlesnake, possesses potent antimicrobial, antitumor, and antifungal properties. Previously, we have shown that its C-terminal fragment, Ctn(15-34), retains the antimicrobial and antitumor activities but is less toxic to healthy cells and has improved serum stability. Here, we investigated the mechanisms of action of Ctn and Ctn(15-34) against Gram-negative bacteria. Both peptides were bactericidal, killing ∼90% of Escherichia coli and Pseudomonas aeruginosa cells within 90-120 and 5-30 min, respectively. Studies of ζ potential at the bacterial cell membrane suggested that both peptides accumulate at and neutralize negative charges on the bacterial surface. Flow cytometry experiments confirmed that both peptides permeabilize the bacterial cell membrane but suggested slightly different mechanisms of action. Ctn(15-34) permeabilized the membrane immediately upon addition to the cells, whereas Ctn had a lag phase before inducing membrane damage and exhibited more complex cell-killing activity, probably because of two different modes of membrane permeabilization. Using surface plasmon resonance and leakage assays with model vesicles, we confirmed that Ctn(15-34) binds to and disrupts lipid membranes and also observed that Ctn(15-34) has a preference for vesicles that mimic bacterial or tumor cell membranes. Atomic force microscopy visualized the effect of these peptides on bacterial cells, and confocal microscopy confirmed their localization on the bacterial surface. Our studies shed light onto the antimicrobial mechanisms of Ctn and Ctn(15-34), suggesting Ctn(15-34) as a promising lead for development as an antibacterial/antitumor agent.


Subject(s)
Anti-Bacterial Agents , Cell Membrane Permeability/drug effects , Cell Membrane , Crotalid Venoms , Crotalus , Escherichia coli , Peptide Fragments , Pseudomonas aeruginosa , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cell Membrane/chemistry , Cell Membrane/metabolism , Crotalid Venoms/chemistry , Crotalid Venoms/pharmacology , Escherichia coli/chemistry , Escherichia coli/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/metabolism , Surface Plasmon Resonance
11.
Front Microbiol ; 8: 775, 2017.
Article in English | MEDLINE | ID: mdl-28522994

ABSTRACT

The increasing prevalence of multidrug-resistant bacteria urges the development of new antibacterial agents. With a broad spectrum activity, antimicrobial peptides have been considered potential antibacterial drug leads. Using bioinformatic tools we have previously shown that viral structural proteins are a rich source for new bioactive peptide sequences, namely antimicrobial and cell-penetrating peptides. Here, we test the efficacy and mechanism of action of the most promising peptides among those previously identified against both Gram-positive and Gram-negative bacteria. Two cell-penetrating peptides, vCPP 0769 and vCPP 2319, have high antibacterial activity against Staphylococcus aureus, MRSA, Escherichia coli, and Pseudomonas aeruginosa, being thus multifunctional. The antibacterial mechanism of action of the two most active viral protein-derived peptides, vAMP 059 and vCPP 2319, was studied in detail. Both peptides act on both Gram-positive S. aureus and Gram-negative P. aeruginosa, with bacterial cell death occurring within minutes. Also, these peptides cause bacterial membrane permeabilization and damage of the bacterial envelope of P. aeruginosa cells. Overall, the results show that structural viral proteins are an abundant source for membrane-active peptides sequences with strong antibacterial properties.

12.
J Med Chem ; 58(21): 8553-63, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26465972

ABSTRACT

In silico dissection of crotalicidin (Ctn), a cathelicidin from a South American pit viper, yielded fragments Ctn[1-14] and Ctn[15-34], which were tested to ascertain to what extent they reproduced the structure and activity of the parent peptide. NMR data showing Ctn to be α-helical at the N-terminus and unstructured at the C-terminus were matched by similar data from the fragments. The peptides were tested against Gram-positive and -negative bacteria and for toxicity against both tumor and healthy cells. Despite its amphipathic α-helical structure, Ctn[1-14] was totally inert toward bacteria or eukaryotic cells. In contrast, unstructured Ctn[15-34] replicated the activity of parent Ctn against Gram-negative bacteria and tumor cells while being significantly less toxic toward eukaryotic cells. This selectivity for bacteria and tumor cells, plus a stability to serum well above that of Ctn, portrays Ctn[15-34] as an appealing candidate for further development as an anti-infective or antitumor lead.


Subject(s)
Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Antineoplastic Agents/chemistry , Crotalid Venoms/chemistry , Gram-Negative Bacteria/drug effects , Amino Acid Sequence , Animals , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents , Antimicrobial Cationic Peptides/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crotalid Venoms/pharmacology , Crotalus , Gram-Negative Bacterial Infections/drug therapy , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Neoplasms/drug therapy , Protein Structure, Secondary , Structure-Activity Relationship , Cathelicidins
SELECTION OF CITATIONS
SEARCH DETAIL
...