Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
PLoS One ; 17(4): e0267164, 2022.
Article in English | MEDLINE | ID: mdl-35421203

ABSTRACT

Extracellular vesicles (EVs) are double membrane structures released by all cell types with identified roles in the generation, transportation, and degradation of amyloid-ß protein (Aß) oligomers in Alzheimer's disease (AD). EVs are thus increasingly recognized to play a neuroprotective role in AD, through their ability to counteract the neurotoxic effects of Aß, possibly through interactions with specific receptors on cell membranes. Our previous studies have identified the amylin receptor (AMY), particularly AMY3 subtype, as a mediator of the deleterious actions of Aß in vitro and in vivo experimental paradigms. In the present study, we demonstrate that AMY3 enriched EVs can bind soluble oligomers of Aß and protect N2a cells against toxic effects of this peptide. The effect was specific to amylin receptor as it was blocked in the presence of amylin receptor antagonist AC253. This notion was supported by reduced Aß binding to EVs from AMY depleted mice compared to those from wild type (Wt) mice. Finally, application of AMY3, but not Wt derived, EVs to hippocampal brain slices improved Aß-induced reduction of long-term potentiation, a cellular surrogate of memory. Collectively, our observations support the role of AMY receptors, particularly AMY3, in EVs as a potential therapeutic target for AD.


Subject(s)
Alzheimer Disease , Extracellular Vesicles , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Extracellular Vesicles/metabolism , Long-Term Potentiation , Mice , Peptide Fragments/toxicity , Receptors, Islet Amyloid Polypeptide/metabolism
3.
Mol Neurobiol ; 58(10): 5369-5382, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34312771

ABSTRACT

Based upon its interactions with amyloid ß peptide (Aß), the amylin receptor, a class B G protein-coupled receptor (GPCR), is a potential modulator of Alzheimer's disease (AD) pathogenesis. However, past pharmacological approaches have failed to resolve whether activation or blockade of this receptor would have greater therapeutic benefit. To address this issue, we generated compound mice expressing a human amyloid precursor protein gene with familial AD mutations in combination with deficiency of amylin receptors produced by hemizygosity for the critical calcitonin receptor subunit of this heterodimeric GPCR. These compound transgenic AD mice demonstrated attenuated responses to human amylin- and Aß-induced depression of hippocampal long-term potentiation (LTP) in keeping with the genetic depletion of amylin receptors. Both the LTP responses and spatial memory (as measured with Morris water maze) in these mice were improved compared to AD mouse controls and, importantly, a reduction in both the amyloid plaque burden and markers of neuroinflammation was observed. Our data support the notion of further development of antagonists of the amylin receptor as AD-modifying therapies.


Subject(s)
Alzheimer Disease/genetics , Maze Learning/physiology , Receptors, Calcitonin/genetics , Receptors, Islet Amyloid Polypeptide/genetics , Spatial Memory/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , Endothelium, Vascular/metabolism , Excitatory Postsynaptic Potentials/physiology , Female , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Receptors, Calcitonin/deficiency , Receptors, Islet Amyloid Polypeptide/deficiency
4.
Sci Rep ; 9(1): 10942, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358858

ABSTRACT

Recent evidence supports involvement of amylin and the amylin receptor in the pathogenesis of Alzheimer's disease (AD). We have previously shown that amylin receptor antagonist, AC253, improves spatial memory in AD mouse models. Herein, we generated and screened a peptide library and identified two short sequence amylin peptides (12-14 aa) that are proteolytically stable, brain penetrant when administered intraperitoneally, neuroprotective against Aß toxicity and restore diminished levels of hippocampal long term potentiation in AD mice. Systemic administration of the peptides for five weeks in aged 5XFAD mice improved spatial memory, reduced amyloid plaque burden, and neuroinflammation. The common residue SQELHRLQTY within the peptides is an essential sequence for preservation of the beneficial effects of the fragments that we report here and constitutes a new pharmacological target. These findings suggest that the amylin receptor antagonism may represent a novel therapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Animals , Female , Hippocampus/drug effects , Islet Amyloid Polypeptide/chemistry , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Receptors, Islet Amyloid Polypeptide/metabolism , Spatial Memory
5.
Commun Biol ; 1: 175, 2018.
Article in English | MEDLINE | ID: mdl-30374465

ABSTRACT

Mechanical signaling involved in molecular interactions lies at the heart of materials science and biological systems, but the mechanisms involved are poorly understood. Here we use nanomechanical sensors and intact human cells to provide unique insights into the signaling pathways of connectivity networks, which deliver the ability to probe cells to produce biologically relevant, quantifiable and reproducible signals. We quantify the mechanical signals from malignant cancer cells, with 10 cells per ml in 1000-fold excess of non-neoplastic human epithelial cells. Moreover, we demonstrate that a direct link between cells and molecules creates a continuous connectivity which acts like a percolating network to propagate mechanical forces over both short and long length-scales. The findings provide mechanistic insights into how cancer cells interact with one another and with their microenvironments, enabling them to invade the surrounding tissues. Further, with this system it is possible to understand how cancer clusters are able to co-ordinate their migration through narrow blood capillaries.

6.
Pharmaceutics ; 10(4)2018 Oct 21.
Article in English | MEDLINE | ID: mdl-30347860

ABSTRACT

Hypoxia can induce chemoresistance, which is a significant clinical obstacle in cancer therapy. Here, we assessed development of hypoxia-induced chemoresistance (HICR) against free versus polymeric cisplatin micelles in a triple negative breast cancer cell line, MDA-MB-231. We then explored two strategies for the modulation of HICR against cisplatin micelles: a) the development of actively targeted micelles; and b) combination therapy with modulators of HICR in MDA-MB-231 cells. Actively targeted cisplatin micelles were prepared through surface modification of acetal-poly(ethylene oxide)-poly(α-carboxyl-ε-caprolactone) (acetal-PEO-PCCL) micelles with epidermal growth factor receptor (EGFR)-targeting peptide, GE11 (YHWYGYTPQNVI). Our results showed that hypoxia induced resistance against free and cisplatin micelles in MDA-MB-231 cells. A significant increase in micellar cisplatin uptake was observed in MDA-MB-231 cells that overexpress EGFR, following surface modification of micelles with GE11. This did not lead to increased cytotoxicity of micellar cisplatin, however. On the other hand, the addition of pharmacological inhibitors of key molecules involved in HICR in MDA-MB-231 cells, i.e., inhibitors of hypoxia inducing factor-1 (HIF-1) and signal transducer and activator of transcription 3 (STAT3), substantially enhanced the cytotoxicity of free and cisplatin micelles. The results indicated the potential benefit of combination therapy with HIF-1 and STAT3 inhibitors in overcoming HICR to free or micellar cisplatin.

7.
J Pharm Pharm Sci ; 21(1): 363-375, 2018.
Article in English | MEDLINE | ID: mdl-30266136

ABSTRACT

Purpose Achieving successful gene therapy requires delivery of a gene vector specifically to the targeted tissue with efficient expression and a good safety profile. The objective of this work was to develop, characterize and determine if a novel gemini surfactant-based lipoplex systems, modified with a cancer-targeting peptide p18-4, could serve this role. Methods The targeting peptide p18-4 was either chemically coupled to a gemini surfactant backbone or physically co-formulated with the lipoplexes. The influence of targeting ligand and formulation strategies on essential physicochemical properties of the lipoplexes was evaluated by dynamic light scattering and small angle X-ray scattering techniques. In vitro transfection activity and cellular toxicity of lipoplexes were assessed in a model human melanoma cell line. Results All lipoplexes zeta potential and particle size were optimal for cellular uptake and physical stability of the system. The lipoplexes adopted an inverted-hexagonal lipid arrangement. The lipoplexes modified with the peptide showed no significant changes in physicochemical properties or lipoplex assembly. The modification of the lipoplexes with the targeting peptide significantly enhanced protein expression 2-6 fold compared to non-modified lipoplexes. In addition, p18-4 modified lipoplexes significantly improved the safety of the lipoplexes. The ability of the p18-4 modified lipoplexes to selectively express the model protein was confirmed by using healthy human epidermal keratinocytes (HEKa). Conclusion The gemini surfactant-based lipoplexes modified with p18-4 peptide showed significantly higher efficiency and safety compared to the system that did not contain a cancer targeting peptide and provided evidence for their potential application to achieve targeted melanoma gene therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Genetic Therapy , Lipids/chemistry , Melanoma/drug therapy , Models, Biological , Peptides/pharmacology , Surface-Active Agents/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Design , Drug Screening Assays, Antitumor , Humans , Melanoma/pathology , Molecular Structure , Particle Size , Peptides/chemistry , Surface Properties , Tumor Cells, Cultured
8.
Alzheimers Dement (N Y) ; 3(1): 44-56, 2017 Jan.
Article in English | MEDLINE | ID: mdl-29067318

ABSTRACT

INTRODUCTION: Amylin receptor serves as a portal for the expression of deleterious effects of amyloid ß-protein (Aß), a key pathologic hallmark of Alzheimer's disease. Previously, we showed that AC253, an amylin receptor antagonist, is neuroprotective against Aß toxicity in vitro and abrogates Aß-induced impairment of hippocampal long-term potentiation. METHODS: Amyloid precursor protein-overexpressing TgCRND8 mice received intracerebroventricularly AC253 for 5 months. New cyclized peptide cAC253 was synthesized and administered intraperitoneally three times a week for 10 weeks in the same mouse model. Cognitive functions were monitored, and pathologic changes were quantified biochemically and immunohistochemically. RESULTS: AC253, when administered intracerebroventricularly, improves spatial memory and learning, increases synaptic integrity, reduces microglial activation without discernible adverse effects in TgCRND8 mice. cAC253 demonstrates superior brain permeability, better proteolytic stability, and enhanced binding affinity to brain amylin receptors after a single intraperitoneal injection. Furthermore, cAC253 administered intraperitoneally also demonstrates improvement in spatial memory in TgCRND8 mice. DISCUSSION: Amylin receptor is a therapeutic target for Alzheimer's disease and represents a disease-modifying therapy for this condition.

9.
J Neuroinflammation ; 14(1): 199, 2017 Oct 06.
Article in English | MEDLINE | ID: mdl-28985759

ABSTRACT

BACKGROUND: Neuroinflammation in the brain consequent to activation of microglia is viewed as an important component of Alzheimer's disease (AD) pathology. Amyloid beta (Aß) protein is known to activate microglia and unleash an inflammatory cascade that eventually results in neuronal dysfunction and death. In this study, we sought to identify the presence of amylin receptors on human fetal and murine microglia and determine whether Aß activation of the inflammasome complex and subsequent release of cytokines is mediated through these receptors. METHODS: The presence of dimeric components of the amylin receptor (calcitonin receptor and receptor activity modifying protein 3) were first immunohistochemically identified on microglia. Purified human fetal microglial (HFM) cultures were incubated with an in vivo microglial marker, DyLight 594-conjugated tomato lectin, and loaded with the membrane-permeant green fluorescent dye, Fluo-8L-AM for measurements of intracellular calcium [Ca2+]i. HFM and BV-2 cells were primed with lipopolysaccharide and then exposed to either human amylin or soluble oligomeric Aß1-42 prior to treatment with and without the amylin receptor antagonist, AC253. Changes in the inflammasome complex, NLRP3 and caspase-1, were examined in treated cell cultures with Western blot and fluorometric assays. RT-PCR measurements were performed to assess cytokine release. Finally, in vivo studies were performed in transgenic mouse model of AD (5xFAD) to examine the effects of systemic administration of AC253 on markers of neuroinflammation in the brain. RESULTS: Acute applications of human amylin or Aß1-42 resulted in an increase in [Ca2+]i that could be blocked by the amylin receptor antagonist, AC253. Activation of the NLRP3 and caspase-1 and subsequent release of cytokines, TNFα and IL-1ß, was diminished by AC253 pretreatment of HFMs and BV2 cells. In vivo, intraperitoneal administration of AC253 resulted in a reduction in microglial markers (Iba-1 and CD68), caspase-1, TNFα, and IL-1ß. These reductions in inflammatory markers were accompanied by reduction in amyloid plaque and size in the brains of 5xFAD mice compared to controls. CONCLUSION: Microglial amylin receptors mediate Aß-evoked inflammation, and amylin receptor antagonists therefore offer an attractive therapeutic target for intervention in AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Inflammation/chemically induced , Microglia/drug effects , Microglia/metabolism , Peptide Fragments/toxicity , Receptors, Islet Amyloid Polypeptide/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Animals , Caspase 1/metabolism , Cell Line, Transformed , Cells, Cultured , Cyclic AMP/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Fetus/cytology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Lipopolysaccharides/toxicity , Male , Mice , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use
10.
Biomaterials ; 144: 17-29, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28818703

ABSTRACT

Traceable poly(ethylene oxide)-poly(ester) micelles were developed through chemical conjugation of a near-infrared (NIR) dye to the poly(ester) end by click chemistry. This strategy was tried for micelles with poly(ε-caprolactone) (PCL) or poly(α-benzyl carboxylate-ε-caprolactone) (PBCL) cores. The surface of both micelles was also modified with the breast cancer targeting peptide, P18-4. The results showed the positive contribution of PBCL over PCL core on micellar thermodynamic and kinetic stability as well as accumulation in primary orthotopic MDA-MB-231 tumors within 4-96 h following intravenous administration in mice. This was in contrast to in vitro studies where better uptake of PEO-PCL versus PEO-PBCL micelles by MDA-MB-231 cells was observed. The presence of P18-4 enhanced the in vitro cell uptake and homing of both polymeric micelles in breast tumors, but only at early time points. In conclusion, the use of developed NIR labeling technique provided means for following the fate of PEO-poly(ester) based nano-carriers in live animals. Our results showed micellar stabilization through the use of PBCL over PCL cores, to have a more significant effect in enhancing the level and duration of nano-carrier accumulation in primary breast tumors than the modification of polymeric micellar surface with breast tumor targeting peptide, P18-4.


Subject(s)
Breast Neoplasms/drug therapy , Drug Delivery Systems , Micelles , Peptides/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Animals , Carbocyanines/administration & dosage , Carbocyanines/pharmacokinetics , Cell Line, Tumor , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/pharmacokinetics , Mice, Nude , Peptides/pharmacokinetics , Polyesters/pharmacokinetics , Polyethylene Glycols/pharmacokinetics
11.
Trends Mol Med ; 23(8): 709-720, 2017 08.
Article in English | MEDLINE | ID: mdl-28694141

ABSTRACT

Alzheimer'sdisease (AD) is a progressive neurodegenerative disorder, characterized by senile plaques constituting extracellular deposits of ß-amyloid (Aß) fibrils. Since Aß accumulation in the brain is considered an early event preceding, by decades, cognitive dysfunction, disease-modifying treatments are aimed at facilitating clearance of this protein from the brain or ameliorating its toxic effects. Recent studies have identified the amylin receptor as a capable mediator of the deleterious actions of Aß and furthermore, administration of amylin receptor-based peptides has been shown to improve spatial memory and learning in transgenic mouse models of AD. Here, by discussing available evidence, we posit that the amylin receptor could be considered a potential therapeutic target for AD, and present the rationale for using amylin receptor antagonists to treat this debilitating condition.


Subject(s)
Alzheimer Disease/drug therapy , Peptides/therapeutic use , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Humans , Mice , Mice, Transgenic , Receptors, Islet Amyloid Polypeptide/genetics
12.
J Med Chem ; 60(12): 4893-4903, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28520410

ABSTRACT

Starting with a previously reported linear breast cancer targeting decapeptide WxEAAYQkFL, here we report the synthesis of a novel cyclic peptide analogue cyclic WXEAAYQkFL. The N- to C-terminus amide cyclized peptide with one d-amino acid (k) displayed higher uptake by breast cancer cells, with minimal uptake by the noncancerous cells compared to the linear peptide with two d-amino acids (x and k), and was stable toward proteolytic degradation. When immobilized on gold microcantilever surface, the cyclic peptide was able to capture breast cancer cells specifically and sense samples with ≥25 cancer cells/mL. Animal studies using mice carrying orthotopic breast MDA-MB-231 tumors showed that the cyclic peptide preferentially accumulates in tumor (2 h after injection) and is rapidly cleared from all other organs except kidneys and liver. The study highlights the discovery of a novel proteolytically stable cyclic peptide that can be used for targeted drug delivery or for enumerating circulating breast tumor cells.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carbocyanines/pharmacokinetics , Cell Line, Tumor , Female , Gold , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Mice, Nude , Peptides, Cyclic/administration & dosage , Proteolysis , Xenograft Model Antitumor Assays
13.
Mol Pharm ; 14(3): 593-604, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28157321

ABSTRACT

The biomarkers or receptors expressed on cancer cells and the targeting ligands with high binding affinity for biomarkers play a key role in early detection and treatment of breast cancer. The breast cancer targeting peptide p160 (12-mer) and its enzymatically stable analogue 18-4 (10-mer) showed marked potential for breast cancer drug delivery using cell studies and animal models. Herein, we used affinity purification, liquid chromatography-tandem mass spectrometry, and proteomics to identify keratin 1 (KRT1) as the target receptor highly expressed on breast cancer cells for p160 peptide(s). Western blot and immunocytochemistry in MCF-7 breast cancer cells confirmed the identity of KRT1. We demonstrate that the p160 or 18-4 binding to MCF-7 breast cancer cells is dependent on the expression of KRT1, and we confirm peptide-KRT1 binding specificity using SPR experiments (Kd ∼ 1.1 µM and 0.98 µM for p160 and 18-4, respectively). Furthermore, we assessed the ability of peptide 18-4 to improve the cellular uptake and anticancer activity of a pro-apoptotic antimicrobial peptide, microcin J25 (MccJ25), in breast cancer cells. A covalent conjugate of peptide 18-4 with MccJ25 showed preferential cytotoxicity toward breast cancer cells with minimal cytotoxicity against normal HUVEC cells. The conjugate inhibited the growth of MDA-MB-435 MDR multidrug-resistant cells with an IC50 comparable to that of nonresistant cells. Conjugation improved selective cellular uptake of MccJ25, and the conjugate triggered cancer cell death by apoptosis. Our findings establish KRT1 as a new marker for breast cancer targeting. Additionally, it pinpoints the potential use of antimicrobial lasso peptides as a novel class of anticancer therapeutics.


Subject(s)
Bacteriocins/pharmacology , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Keratin-1/metabolism , Peptides/pharmacology , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Cell Line , Cell Line, Tumor , Drug Delivery Systems/methods , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Human Umbilical Vein Endothelial Cells , Humans , Ligands , MCF-7 Cells
15.
Sensors (Basel) ; 15(8): 19429-42, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26262621

ABSTRACT

The adsorption of the proteins CD13, mucin and bovine serum albumin on VLGXE-Au and YNGRT-Au interfaces was monitored by electrochemical impedance spectroscopy in the presence of [Fe(CN)6](3-/4-). The hydrophobicity of the Au surface was tailored using specific peptides, blocking agents and diluents. The combination of blocking agents (ethanolamine or n-butylamine) and diluents (hexanethiol or 2-mercaptoethanol) was used to prepare various peptide-modified Au surfaces. Protein adsorption onto the peptide-Au surfaces modified with the combination of n-butylamine and hexanethiol produced a dramatic decrease in the charge transfer resistance, Rct, for all three proteins. In contrast, polar peptide-surfaces induced a minimal change in Rct for all three proteins. Furthermore, an increase in Rct was observed with CD13 (an aminopeptidase overexpressed in certain cancers) in comparison to the other proteins when the VLGXE-Au surface was modified with n-butylamine as a blocking agent. The electrochemical data indicated that protein adsorption may be modulated by tailoring the peptide sequence on Au surfaces and that blocking agents and diluents play a key role in promoting or preventing protein adsorption. The peptide-Au platform may also be used for targeting cancer biomarkers with designer peptides.


Subject(s)
Electrochemistry/methods , Gold/chemistry , Peptides/chemistry , Serum Albumin, Bovine/isolation & purification , Adsorption , Animals , Cattle , Electric Impedance , Hydrophobic and Hydrophilic Interactions , Immobilized Proteins/chemistry , Protein Binding , Solutions , Surface Properties
16.
Methods Mol Biol ; 1248: 239-47, 2015.
Article in English | MEDLINE | ID: mdl-25616337

ABSTRACT

The identification of cancer cell-specific ligands is a key requirement for the targeted delivery of chemotherapeutic agents. Usually phage display system is employed to discover cancer-specific peptides through a biopanning process. Synthetic peptide array libraries can be used as a complementary method to phage display for screening and identifying cancer cell-specific ligands. Here, we describe a peptide array-whole cell binding assay to identify cancer cell-specific peptides. A peptide array library based on a lead dodecapeptide, p160, is synthesized on a functionalized cellulose membrane using solid phase chemistry and a robotic synthesizer. The relative binding affinity of the peptide library is evaluated by incubating the library with fluorescently labeled cancerous or non-cancerous cells. Thereby the assay allows picking peptides that show selective and high binding to cancerous cells. These peptides represent potential candidates for use in cancer-targeted drug delivery, imaging, and diagnosis.


Subject(s)
Drug Delivery Systems , Neoplasms , Oligopeptides , Peptide Library , Animals , Cell Line, Tumor , Humans , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/metabolism , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/pharmacology , Protein Binding
17.
J Med Chem ; 56(19): 7564-73, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24028446

ABSTRACT

The efficacy of chemotherapeutic doxorubucin (Dox) in cancer treatment is limited by two main factors, nonspecific toxicity and the emergence of tumor resistance. To overcome these hurdles, in this study peptide-Dox conjugates were prepared. A decapeptide 18-4a (NH2-WxEAAYQkFL-CONH2) [corrected] with high specificity for breast cancer cells and improved proteolytic stability was conjugated to Dox to give peptide-Dox ester (1) and amide (2) conjugates. Cell uptake studies showed that the conjugates were 6-10 times selective for breast cancerous cells (MCF-7 and MDA-MB-435) over noncancerous cells (HUVECs and MCF-10A). Conjugate 1 displayed similar toxicity as free Dox toward the breast cancerous cells and was about 40 times less toxic toward the noncancerous cells and 4-fold more toxic toward the Dox resistant MDA-MB-435-MDR cells than the free Dox. These data suggest that conjugate 1 can be used as a potential prodrug for improving the therapeutic index of Dox and potentially many other cytotoxic drugs.


Subject(s)
Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Doxorubicin/analogs & derivatives , Doxorubicin/chemistry , Oligopeptides/chemistry , Prodrugs/chemistry , Antineoplastic Agents/pharmacology , Cell Line , Cell Line, Tumor/drug effects , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , Female , Humans , Oligopeptides/pharmacology , Prodrugs/pharmacology , Structure-Activity Relationship , Verapamil/pharmacology
18.
Bioorg Med Chem ; 21(13): 3715-22, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23673216

ABSTRACT

Class IIa bacteriocins are potent antimicrobial peptides produced by lactic acid bacteria to destroy competing microorganisms. The N-terminal domain of these peptides consists of a conserved YGNGV sequence and a disulphide bond. The YGNGV motif is essential for activity, whereas, the two cysteines involved in the disulphide bond can be replaced with hydrophobic residues. The C-terminal region has variable sequences, and folds into a conserved amphipathic α-helical structure. To elucidate the structure-activity relationship in the N-terminal domain of these peptides, three analogues (1-3) of a class IIa bacteriocin, Leucocin A (LeuA), were designed and synthesized by replacing the N-terminal ß-sheet residues of the native peptide with shorter ß-turn motifs. Such replacement abolished the antibacterial activity in the analogues, however, analogue 1 was able to competitively inhibit the activity of native LeuA. Native LeuA (37-mer) was synthesized using native chemical ligation method in high yield. Solution conformation study using circular dichroism spectroscopy and molecular dynamics simulations suggested that the C-terminal region of analogue 1 adopts helical folding as found in LeuA, while the N-terminal region did not fold into ß-sheet conformation. These structure-activity studies highlight the role of proper folding and complete sequence in the activity of class IIa bacteriocins.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteriocins/chemistry , Bacteriocins/pharmacology , Amino Acid Sequence , Anti-Bacterial Agents/chemical synthesis , Bacteriocins/chemical synthesis , Carnobacteriaceae/drug effects , Humans , Listeria monocytogenes/drug effects , Listeriosis/drug therapy , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Structural Homology, Protein , Structure-Activity Relationship
19.
Biomaterials ; 34(16): 4089-4097, 2013 May.
Article in English | MEDLINE | ID: mdl-23465829

ABSTRACT

This study aimed to develop actively targeted liposomal formulations of doxorubicin (DOX) using an engineered breast tumor targeting peptide ligand, p18-4 (WxEAAYQrFL). Towards this goal, stealth liposomes bearing different molar ratios of p18-4 peptide (1.5 and 0.3 peptide/total lipid mol %), namely HD and LD liposomes, were successfully prepared. The effect of p18-4 peptide modification and density on breast cancer cell uptake, selective cytotoxicity as well as inhibition of tumor growth and the tissue disposition of encapsulated DOX in breast tumor xenograft models in mice were assessed. The results showed a 2.4 and 5 folds decrease in the IC50 of HD liposomes in MDA-MB-435 and MCF-7 breast tumor cells, respectively. Although LD liposomes showed less (1.6 and 2.2 folds) decrease in the IC50 of DOX in the same breast cancer cell lines, they were more selective in their cytotoxic effect and uptake towards breast cancer over normal breast epithelial cells, MCF10A. Evaluation of the anticancer activity in NOD-SCID mice bearing MDA-MB-435 xenografts after receiving six i.v. injections of 2.5mg/kg/week DOX equivalent showed a superior anticancer activity for LD liposomal DOX compared to HD and unmodified liposomal formulations. Mice treated with LD liposomal DOX illustrated 4.8 folds reduction in the mean relative tumor volume compared to non-targeted DOX liposomes. This was despite similar tumor accumulation of DOX as part of LD liposomes compared to that for unmodified liposomes 24h following the last injection. In contrast, HD liposomes showed decreased DOX accumulation in the tumor and preferential uptake by liver and spleen. Treatment with unmodified and LD liposomes did not have any adverse impact on the activity level and mean body weight of live animals during the study period. In conclusion, surface modification of liposomal DOX with engineered p18-4 peptide at an optimum density can improve the antitumor efficacy and selectivity of liposomal DOX.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Delivery Systems , Peptides/pharmacology , Protein Engineering , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Body Weight/drug effects , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Female , Ligands , Mice , Mice, SCID , Molecular Sequence Data , Peptides/chemistry , Tissue Distribution/drug effects , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
20.
Cancer Lett ; 334(2): 284-92, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23073474

ABSTRACT

Chemotherapy is still the treatment of choice for many types of cancer; but its effectiveness is hampered by dose limiting toxicity. Properly designed delivery systems can overcome this shortcoming by reducing the non-specific distribution and toxicity of chemotherapeutics in healthy organs and at the same time increasing drug concentrations at tumor tissue. In this study, we developed stealth liposomal formulations of doxorubicin (DOX) having a novel stable engineered peptide ligand, namely p18-4, that binds specifically to breast cancer cell line MDA-MB-435 on its surface. The coupling of p18-4 to liposomes was carried out through conventional, post insertion and post conjugation techniques and prepared liposomes were characterized for their size and level of peptide modification. The p18-4 decorated liposomal DOX formulations were then evaluated for their cellular uptake as well as cytotoxicity against the human breast cancer MDA-MB-435 cells. In this context, the effect of coupling technique on the uptake and cytotoxicity of p18-4 liposomal DOX in MDA-MB-435 cells was evaluated. The conventional and post conjugation methods of peptide incorporation were found to be more reliable for the preparation of p18-4 decorated liposomes for active DOX targeting to MDA-MB-435 cells. p18-4 decoration of liposomes by these methods did not have a notable effect on the size of prepared liposomes and DOX release, but increased the uptake and cytotoxicity of encapsulated DOX in MDA-MB-435 cells. The results show a potential for p18-4 decorated liposomes prepared by conventional and post conjugation method for tumor targeted delivery of DOX in breast tumor models.


Subject(s)
Breast Neoplasms/drug therapy , Doxorubicin/analogs & derivatives , Drug Delivery Systems/methods , Liposomes/administration & dosage , Peptides/administration & dosage , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Breast Neoplasms/pathology , Cell Line, Tumor , Chemistry, Physical , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Female , Genetic Engineering/methods , Humans , Liposomes/chemistry , Particle Size , Peptides/chemistry , Peptides/genetics , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...