Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(34): e2202926119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35969786

ABSTRACT

The Ca2+-activated SK4 K+ channel is gated by Ca2+-calmodulin (CaM) and is expressed in immune cells, brain, and heart. A cryoelectron microscopy (cryo-EM) structure of the human SK4 K+ channel recently revealed four CaM molecules per channel tetramer, where the apo CaM C-lobe and the holo CaM N-lobe interact with the proximal carboxyl terminus and the linker S4-S5, respectively, to gate the channel. Here, we show that phosphatidylinositol 4-5 bisphosphate (PIP2) potently activates SK4 channels by docking to the boundary of the CaM-binding domain. An allosteric blocker, BA6b9, was designed to act to the CaM-PIP2-binding domain, a previously untargeted region of SK4 channels, at the interface of the proximal carboxyl terminus and the linker S4-S5. Site-directed mutagenesis, molecular docking, and patch-clamp electrophysiology indicate that BA6b9 inhibits SK4 channels by interacting with two specific residues, Arg191 and His192 in the linker S4-S5, not conserved in SK1-SK3 subunits, thereby conferring selectivity and preventing the Ca2+-CaM N-lobe from properly interacting with the channel linker region. Immunohistochemistry of the SK4 channel protein in rat hearts showed a widespread expression in the sarcolemma of atrial myocytes, with a sarcomeric striated Z-band pattern, and a weaker occurrence in the ventricle but a marked incidence at the intercalated discs. BA6b9 significantly prolonged atrial and atrioventricular effective refractory periods in rat isolated hearts and reduced atrial fibrillation induction ex vivo. Our work suggests that inhibition of SK4 K+ channels by targeting drugs to the CaM-PIP2-binding domain provides a promising anti-arrhythmic therapy.


Subject(s)
Atrial Fibrillation , Calmodulin , Intermediate-Conductance Calcium-Activated Potassium Channels , Potassium Channel Blockers , Animals , Atrial Fibrillation/drug therapy , Calcium Signaling , Calmodulin/metabolism , Cryoelectron Microscopy , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Molecular Docking Simulation , Mutagenesis, Site-Directed , Phosphatidylinositol 4,5-Diphosphate , Potassium Channel Blockers/pharmacology , Rats
2.
Angew Chem Int Ed Engl ; 61(29): e202205368, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35536102

ABSTRACT

LPtIV F(Aryl) complexes bearing a bulky bidentate 2-[bis(adamant-1-yl)phosphino]phenoxide ligand (L) demonstrate excellent reactivity and selectivity in the arylation of X-H (X=S, N) bonds of amino acid residues in unprotected peptides under mild, including aqueous, conditions. Stepwise addition of these complexes allowed a convenient one-pot introduction of different aromatic groups in the X-H bonds of Cys and N terminus. PtIV reagents can also be used to further arylate N-H bonds in Lys and Trp providing access to peptides bearing multiple aromatic groups.


Subject(s)
Peptides , Indicators and Reagents , Ligands , Peptides/chemistry
3.
Sci Adv ; 8(20): eabn1171, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35584224

ABSTRACT

Isoprenoids are synthesized by the prenyltransferase superfamily, which is subdivided according to the product stereoisomerism and length. In short- and medium-chain isoprenoids, product length correlates with active site volume. However, enzymes synthesizing long-chain products and rubber synthases fail to conform to this paradigm, because of an unexpectedly small active site. Here, we focused on the human cis-prenyltransferase complex (hcis-PT), residing at the endoplasmic reticulum membrane and playing a crucial role in protein glycosylation. Crystallographic investigation of hcis-PT along the reaction cycle revealed an outlet for the elongating product. Hydrogen-deuterium exchange mass spectrometry analysis showed that the hydrophobic active site core is flanked by dynamic regions consistent with separate inlet and outlet orifices. Last, using a fluorescence substrate analog, we show that product elongation and membrane association are closely correlated. Together, our results support direct membrane insertion of the elongating isoprenoid during catalysis, uncoupling active site volume from product length.

4.
Int J Mol Sci ; 22(17)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34502483

ABSTRACT

We examined the effects of ALOS4, a cyclic peptide discovered previously by phage library selection against integrin αvß3, on a human melanoma (A375) xenograft model to determine its abilities as a potential anti-cancer agent. We found that ALOS4 promoted healthy weight gain in A375-engrafted nude mice and reduced melanoma tumor mass and volume. Despite these positive changes, examination of the tumor tissue did not indicate any significant effects on proliferation, mitotic index, tissue vascularization, or reduction of αSMA or Ki-67 tumor markers. Modulation in overall expression of critical downstream αvß3 integrin factors, such as FAK and Src, as well as reductions in gene expression of c-Fos and c-Jun transcription factors, indirectly confirmed our suspicions that ALOS4 is likely acting through an integrin-mediated pathway. Further, we found no overt formulation issues with ALOS4 regarding interaction with standard inert laboratory materials (polypropylene, borosilicate glass) or with pH and temperature stability under prolonged storage. Collectively, ALOS4 appears to be safe, chemically stable, and produces anti-cancer effects in a human xenograft model of melanoma. We believe these results suggest a role for ALOS4 in an integrin-mediated pathway in exerting its anti-cancer effects possibly through immune response modulation.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma, Experimental/drug therapy , Peptides, Cyclic/pharmacology , Animals , Cell Line, Tumor , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred ICR , Mice, Nude , Xenograft Model Antitumor Assays
5.
Angew Chem Int Ed Engl ; 60(31): 17164-17170, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34014019

ABSTRACT

The structural arrangement of amino acid residues in native enzymes underlies their remarkable catalytic properties, thus providing a notable point of reference for designing potent yet simple biomimetic catalysts. Herein, we describe a minimalistic approach to construct a dipeptide-based nano-superstructure with enzyme-like activity. The self-assembled biocatalyst comprises one peptide as a single building block, readily synthesized from histidine. Through coordination with zinc ion, the peptide self-assembly procedure allows the formation of supramolecular ß-sheet ordered nanocrystals, which can be used as basic units to further construct higher-order superstructure. As a result, remarkable hydrolysis activity and enduring stability are demonstrated. Our work exemplifies the use of a bioinspired supramolecular assembly approach to develop next-generation biocatalysts for biotechnological applications.


Subject(s)
Nanoparticles/chemistry , Peptides/chemistry , Histidine/chemistry , Hydrolysis , Particle Size , Peptides/chemical synthesis
6.
Oncotarget ; 8(1): 757-768, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27768593

ABSTRACT

The newly discovered short (9 amino acid) non-RGD S-S bridged cyclic peptide ALOS-4 (H-cycl(Cys-Ser-Ser-Ala-Gly-Ser-Leu-Phe-Cys)-OH), which binds to integrin αvß3 is investigated as peptide carrier for targeted drug delivery against human metastatic melanoma. ALOS4 binds specifically the αvß3 overexpressing human metastatic melanoma WM-266-4 cell line both in vitro and in ex vivo assays. Coupling ALOS4 to the topoisomerase I inhibitor Camptothecin (ALOS4-CPT) increases the cytotoxicity of CPT against human metastatic melanoma cells while reduces dramatically the cytotoxicity against non-cancerous cells as measured by the levels of γH2A.X, active caspase 3 and cell viability. Moreover, conjugating ALOS4 to CPT even increases the chemo-stability of CPT under physiological pH. Bioinformatic analysis using Rosetta platform revealed potential docking sites of ALOS4 on the αvß3 integrin which are distinct from the RGD binding sites. We propose to use this specific non-RGD cyclic peptide as the therapeutic carrier for conjugation of drugs in order to improve efficacy and reduce toxicity of currently available treatments of human malignant melanoma.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Discovery , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Stability , Humans , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Mice , Models, Molecular , Molecular Conformation , Molecular Structure , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptides, Cyclic/chemical synthesis , Protein Binding , Xenograft Model Antitumor Assays
7.
Oncotarget ; 7(39): 63549-63560, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27556860

ABSTRACT

ALOS4, a unique synthetic cyclic peptide without resemblance to known integrin ligand sequences, was discovered through repeated biopanning with pIII phage expressing a disulfide-constrained nonapeptide library. Binding assays using a FITC-labeled analogue demonstrated selective binding to immobilized αvß3 and a lack of significant binding to other common proteins, such as bovine serum albumin and collagen. In B16F10 cell cultures, ALOS4 treatment at 72 h inhibited cell migration (30%) and adhesion (up to 67%). Immunofluorescent imaging an ALOS4-FITC analogue with B16F10 cells demonstrated rapid cell surface binding, and uptake and localization in the cytoplasm. Daily injections of ALOS4 (0.1, 0.3 or 0.5 mg/kg i.p.) to mice inoculated with B16F10 mouse melanoma cells in two different cancer models, metastatic and subcutaneous tumor, resulted in reduction of lung tumor count (metastatic) and tumor mass (subcutaneous) and increased survival of animals monitored to 45 and 60 days, respectively. Examination of cellular activity indicated that ALOS4 produces inhibition of cell migration and adhesion in a concentration-dependent manner. Collectively, these results suggest that ALOS4 is a structurally-unique selective αvß3 integrin ligand with potential anti-metastatic activity.


Subject(s)
Integrin alphaVbeta3/metabolism , Lung Neoplasms/prevention & control , Melanoma, Experimental/prevention & control , Peptides, Cyclic/pharmacology , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Peptide Library , Tumor Cells, Cultured
8.
Biopolymers ; 106(1): 119-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26662352

ABSTRACT

Compact carriers for peptidyl delivery systems (PDSs) loaded with various drugs were synthesized using a simple and convenient solid phase organic synthesis strategy, including semi-orthogonal functional group protection schemes. Each attachment point of the compact carrier can thus be bound to an anticancer agent through a biodegradable covalent link. Chemo- and biostability experiments of a model peptidyl platform loaded with three different drugs revealed pH and liver homogenate (metabolic) dependent sequential release behavior. The versatility of this approach will serve to expedite the preparation of PDS libraries. This approach may prove useful for applications suitable for personalized medicine where multiple drug delivery is required in a sequential and controlled fashion.


Subject(s)
Drug Delivery Systems , Peptides/administration & dosage , Cell Line , Chromatography, High Pressure Liquid , Humans , Magnetic Resonance Spectroscopy , Spectrometry, Mass, Electrospray Ionization
9.
Biopolymers ; 104(6): 743-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26058565

ABSTRACT

Peptide conjugates containing somatostatin (SST) cyclic analogs as a targeting moiety are able to deliver chemotherapeutic agents specifically to cancer cells expressing SST receptors (SSTRs), and hence increasing their local efficacy while limiting the peripheral toxicity. Here, we report on the synthesis and biochemical characterization of new SSTR-specific anticancer peptide conjugates, with different anticancer payloads acting through different oncogenic mechanisms to evaluate their biological activities and to provide a comparative study of their drug release profiles. The SSTR2-specific backbone cyclic peptide 3207-86 was chosen for the synthesis of a variety of novel anticancer drug conjugates with a broad drug release capabilities. The N-terminus of 3207-86 was equipped with GABA to generate free amino group available for the conjugation of chlorambucil, Camptothecin (CPT), Combretastatin 4A, ABT-751, and Amonafide through the formation of various biodegradable bonds. The chemo- and biostability/drug release of all the synthetic compounds was investigated at various pHs and in the presence of mouse liver homogenate, respectively. Their selective cytotoxic effect was evaluated on several human cancer cell lines that overexpress SSTR2. Compared with the free drugs, our peptide-drug conjugates exhibited considerable cytotoxic effect on cancer cell lines versus low SSTR2-expressed human embryonic kidney cells. Functional versatility of the conjugates was reflected in the variability of their drug release profiles, whereas the conserved sequence of a selective binding to the SSTR2 likely preserved their binding to the receptor and consequently their favorable toxicity toward targeted cancer cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Somatostatin/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cyclization , Drug Delivery Systems , Humans , Mice , Somatostatin/therapeutic use , Spectrometry, Mass, Electrospray Ionization
10.
Med Chem ; 11(4): 373-82, 2015.
Article in English | MEDLINE | ID: mdl-25524547

ABSTRACT

We tested the antiproliferative activity and mechanism of the action of several novel aminoacridine derivatives. Six different cancer cell lines were used to evaluate the potential cytotoxic effect of eleven aminoacridine-based molecules. A standard MTT assay was used for cell bioavailability analysis. Additionally, the potential cytotoxic effect of the tested compounds on non-cancer cells was investigated in rat skeletal muscle myotubes (L6) and in bovine aortic smooth muscle cells. In order to investigate whether the DNA binding activity of tested compounds correlated with their cytotoxic effect, circular dichroism (CD) measurement and DNA T4 ligase assay were performed. Finally, the potential mutagenic activity of the lead compound 5 was investigated. The cytotoxic effect of compound 5 in cancer cells was obtained in lower concentrations than the well-known: 9- aminoacridine based drug, amsacrine. The lead compound binds to DNA, but in a different mode than the parent molecules. Additionally, compound 5 was not cytotoxic in the effective range of concentrations in non-cancer cells. In identical concentrations, the parent compound (9-aminoacridine) and amsacrine were extremely toxic for both types of these normal cells. Finally, based on CD measurement and T4 ligase assay, it was confirmed that 5 binds to DNA but in different from the parent compounds manner. Important to mention, that compound 5 might have increased mutagenic activity which must be verified in vivo. Based on these in vitro results, we conclude that 5 is a more potent and more selective antiprolifirative compound than amsacrine. Compound 5 was also more effective in HepG2 and P-12 cells. Thus, 5 is suitable for future in vivo biological evaluation and its structure might be used as a basis for developing novel anticancer drugs.


Subject(s)
Aminoacridines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Intercalating Agents/pharmacology , Aminoacridines/pharmacology , Amsacrine/chemistry , Amsacrine/toxicity , Animals , Antineoplastic Agents/pharmacology , Cattle , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA/antagonists & inhibitors , DNA/chemistry , DNA Ligase ATP , DNA Ligases/chemistry , Humans , Intercalating Agents/chemistry , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Organ Specificity , Rats , Structure-Activity Relationship
11.
Eur J Med Chem ; 85: 139-46, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25084142

ABSTRACT

Bi-nuclear amino acid platforms loaded with various drugs for conjugation to a peptide carrier were synthesized using simple and convenient orthogonally protective solid-phase organic synthesis (SPOS). Each arm of the platform carries a different anticancer agent linked through the same or different functional group, providing discrete chemo- and bio-release profiles for each drug, and also enabling "switch off/switch on" regulation of drug cytotoxicity by conjugation to the platform and to a cell targeting peptide. The versatility of this approach enables efficient production of drug-loaded platforms and determination of favorable drug combinations/modes of linkage for subsequent conjugation to a carrier moiety for targeted cancer cell therapy. The results presented here potentiate the application of amino acid platforms for targeted drug delivery (TDD).


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Carriers/chemistry , Amino Acids/chemistry , Animals , Antineoplastic Agents/toxicity , Cell Line, Tumor , Drug Carriers/chemical synthesis , Drug Carriers/metabolism , Drug Stability , Humans , Liver/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...