Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Publication year range
1.
Biopolymers ; 104(4): 384-394, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25670241

ABSTRACT

Tryptophan (Trp) is a naturally occurring amino acid, which exhibits fluorescence emission properties that are dependent on the polarity of the local environment around the Trp side chain. However, this sensitivity also complicates interpretation of fluorescence emission data. A non-natural analogue of tryptophan, ß-(1-azulenyl)-L-alanine, exhibits fluorescence insensitive to local solvent polarity and does not impact the structure or characteristics of several peptides examined. In this study, we investigated the effect of replacing Trp with ß-(1-azulenyl)-L-alanine in the well-known bee-venom peptide melittin. This peptide provides a model framework for investigating the impact of replacing Trp with ß-(1-azulenyl)-L-alanine in a functional peptide system that undergoes significant shifts in Trp fluorescence emission upon binding to lipid bilayers. Microbiological methods including assessment of the antimicrobial activity by minimal inhibitory concentration (MIC) assays and bacterial membrane permeability assays indicated little difference between the Trp and the ß-(1-azulenyl)-L-alanine-substituted versions of melittin. Circular dichroism spectroscopy showed both that peptides adopted the expected α-helical structures when bound to phospholipid bilayers and electrophysiological analysis indicated that both created membrane disruptions leading to significant conductance increases across model membranes. Both peptides exhibited a marked protection of the respective fluorophores when bound to bilayers indicating a similar membrane-bound topology. As expected, while fluorescence quenching and CD indicate the peptides are stably bound to lipid vesicles, the peptide containing ß-(1-azulenyl)-L-alanine exhibited no fluorescence emission shift upon binding while the natural Trp exhibited >10 nm shift in emission spectrum barycenter. Taken together, the ß-(1-azulenyl)-L-alanine can serve as a solvent insensitive alternative to Trp that does not have significant impacts on structure or function of membrane interacting peptides.


Subject(s)
Fluorescence , Lipid Bilayers/chemistry , Melitten , Tryptophan , Melitten/analogs & derivatives , Melitten/chemistry , Protein Structure, Secondary , Tryptophan/analogs & derivatives , Tryptophan/chemistry
2.
Amino Acids ; 47(7): 1433-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25868656

ABSTRACT

Conjugate compounds constitute a new class of molecules of important biological interest mainly for the treatment of diseases such as cancer. The N-terminus region of cationic peptides has been described as important for their biological activity. The aim of this study was to evaluate the lytic peptide Hecate (FALALKALKKALKKLKKALKKAL) and the effect of conjugating this macromolecule with gallic acid (C7H6O5) in terms of structure, anti-cancer activity, and toxicity. An N-terminus GA-Hecate peptide conjugate was synthesized to provide information regarding the relationship between the amino-terminal region and its charge and the secondary structure and biological activity of the peptide; and the effects of gallic acid on these parameters. Peptide secondary structure was confirmed using circular dichroism (CD). The CD measurements showed that the peptide has a high incidence of α-helical structures in the presence of SDS and LPC, while GA-Hecate presented lower incidence of α-helical structures in the same chemical environment. An evaluation of the anti-cancer activity in HeLa cancer cells indicated that both peptides are active, but that coupling gallic acid at the N-terminus decreased the activity of the free peptide. GA-Hecate showed lower activity in non-tumor keratinocyte cells but higher hemolytic activity. Our findings suggest that the N-terminus of Hecate plays an important role in its activity against cervical cancer by affecting it secondary structure, toxicity, and hemolytic activity. This study highlights the importance of the N-terminus in antitumor activity and could provide an important tool for developing new anti-cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Gallic Acid/pharmacology , Hemolytic Agents/pharmacology , Melitten/analogs & derivatives , Amino Acid Sequence , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Erythrocytes/drug effects , Female , HeLa Cells , Humans , Melitten/pharmacology , Molecular Sequence Data , Uterine Cervical Neoplasms
3.
J Am Chem Soc ; 134(30): 12732-41, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22731650

ABSTRACT

We recently developed an orthogonal, high-throughput assay to identify peptides that self-assemble into potent, equilibrium pores in synthetic lipid bilayers. Here, we use this assay as a high-throughput screen to select highly potent pore-forming peptides from a 7776-member rational combinatorial peptide library based on the sequence of the natural pore-forming peptide toxin melittin. In the library we varied ten critical residues in the melittin sequence, chosen to test specific structural hypotheses about the mechanism of pore formation. Using the new high-throughput assay, we screened the library for gain-of-function sequences at a peptide to lipid ratio of 1:1000 where native melittin is not active. More than 99% of the library sequences were also inactive under these conditions. A small number of library members (0.1%) were highly active. From these we identified 14 potent, gain-of-function, pore-forming sequences. These sequences differed from melittin in only 2-6 amino acids out of 26. Some native residues were highly conserved and others were consistently changed. The two factors that were essential for gain-of-function were the preservation of melittin's proline-dependent break in the middle of the helix and the improvement and extension the amphipathic nature of the α-helix. In particular the highly cationic carboxyl-terminal sequence of melittin, is consistently changed in the gain-of-function variants to a sequence that it is capable of participating in an extended amphipathic α-helix. The most potent variants reside in a membrane-spanning orientation, in contrast to the parent melittin, which is predominantly surface bound. This structural information, taken together with the high-throughput tools developed for this work, enable the identification, refinement and optimization of pore-forming peptides for many potential applications.


Subject(s)
Melitten/analogs & derivatives , Melitten/pharmacology , Unilamellar Liposomes/metabolism , Amino Acid Sequence , Conserved Sequence , High-Throughput Screening Assays , Molecular Sequence Data , Peptide Library , Phospholipids/metabolism
4.
Carbohydr Polym ; 280: 119033, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35027135

ABSTRACT

Bacteria-induced wound infections and multifunctional hydrogels have received widespread attention in wound repair. In this study, self-assembling peptides (SAPs) were grafted on O-carboxymethyl chitosan (O-CMCS), and compact spatial structure and good drug sustained-release effect on mel-d1, a new AMP designed based on melittin with the same antimicrobial activity but lower cytotoxicity and ciprofloxacin (CIP) were obtained. In vivo test showed that the O-CMCS/SAP hydrogel loaded with CIP and mel-d1 accelerated the wound closure speed caused by infection of Escherichia coli and skin tissue regeneration. Both of the enhanced interaction between O-CMCS/SAP and CIP/Mel-d1 because of the hydrophobic interaction and π-π stacking, and the potential tissue healing ability of SAP played important roles. This study provided a rational design method of O-CMCS by grafting SAPs to give a wider range of biological functions.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bandages , Chitosan/analogs & derivatives , Ciprofloxacin/pharmacology , Escherichia coli Infections/therapy , Melitten/analogs & derivatives , Wound Healing , Animals , Cell Membrane/drug effects , Delayed-Action Preparations , Drug Design , Escherichia coli/drug effects , Hydrogels , Hydrophobic and Hydrophilic Interactions , Male , Melitten/chemistry , Melitten/pharmacology , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Peptides/pharmacology , Rheology
5.
Biochim Biophys Acta ; 1798(2): 128-39, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19635451

ABSTRACT

Melittin, the major component of the honey bee venom, is a 26-residue hemolytic and membrane active peptide. Structures of melittin determined either in lipid environments by NMR or by use of X-ray demonstrated two helical regions at the N- and C-termini connected by a hinge or a bend at the middle. Here, we show that deletion of the hinge residues along with two C-terminal terminal Gln residues (Q25 and Q26), yielding a peptide analog of 19-residue or Mel-H, did not affect antibacterial activity but resulted in a somewhat reduced hemolytic activity. A diastereomer of Mel-H or Mel-(d)H containing d-amino acids [(d)V5, (d)V8, (d)L11 and (d)K16] showed further reduction in hemolytic activity without lowering antibacterial activity. We have carried out NMR structures, dynamics (H-D exchange and proton relaxation), membrane localization by spin labeled lipids, pulse-field-gradient (PFG) NMR and isothermal titration calorimetry (ITC) in dodecylphosphocholine (DPC) micelles, as a mimic to eukaryotic membrane, to gain insights into cell selectivity of these melittin analogs. PFG-NMR showed Mel-H and Mel-(d)H both were similarly partitioned into DPC micelles. ITC demonstrated that Mel-H and Mel-(d)H interact with DPC with similar affinity. The micelle-bound structure of Mel-H delineated a straight helical conformation, whereas Mel-(d)H showed multiple beta-turns at the N-terminus and a short helix at the C-terminus. The backbone amide-proton exchange with solvent D(2)O demonstrated a large difference in dynamics between Mel-H and Mel-(d)H, whereby almost all backbone protons of Mel-(d)H showed a much faster rate of exchange as compared to Mel-H. Proton T(1) relaxation had suggested a mobile backbone of Mel-(d)H peptide in DPC micelles. Resonance perturbation by paramagnetic lipids indicated that Mel-H inserted deeper into DPC micelles, whereas Mel-(d)H is largely located at the surface of the micelle. Taken together, results presented in this study demonstrated that the poor hemolytic activity of the d-amino acid containing analogs of antimicrobial peptides may be correlated with their flexible dynamics at the membrane surface.


Subject(s)
Erythrocytes/chemistry , Melitten/chemistry , Micelles , Animals , Erythrocytes/metabolism , Hemolysis , Humans , Melitten/analogs & derivatives , Melitten/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary/physiology , Structure-Activity Relationship
6.
Drug Discov Today ; 26(4): 1053-1059, 2021 04.
Article in English | MEDLINE | ID: mdl-33450177

ABSTRACT

Melittin is vital for the endosomal escape of nanoparticles, but its excessive cytotoxicity in mammalian cells limits its value as a potential therapeutic agent. Several novel analogs of melittin have been optimized and characterized to establish a non-toxic melittin-based gene delivery system, in which the sequences of the melittin peptides were altered to reduce their cytotoxic activity. This review focusses on the involvement of melittin in nanoparticle endosomal escape and on the construction of melittin conjugates to boost gene delivery. Endosomal escape mechanisms for melittin, as well as the development of melittin as a therapeutic agent and its potential applications in nanomedicine, are discussed.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Melitten , Nanoparticles/therapeutic use , Antimicrobial Peptides/pharmacology , Endosomes/physiology , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Melitten/analogs & derivatives , Melitten/pharmacology
7.
ACS Appl Mater Interfaces ; 13(36): 42533-42542, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34472829

ABSTRACT

Despite potency against a variety of cancers in preclinical systems, melittin (MEL), a major peptide in bee venom, exhibits non-specific toxicity, severe hemolytic activity, and poor pharmacological properties. Therefore, its advancement in the clinical translation system has been limited to early-stage trials. Herein, we report a biohybrid involving a bottlebrush-architectured poly(ethylene glycol) (PEG) and MEL. Termed pacMEL, the conjugate consists of a high-density PEG arrangement, which provides MEL with steric inhibition against protein access, while the high molecular weight of pacMEL substantially enhances plasma pharmacokinetics with a ∼10-fold increase in the area under the curve (AUC∞) compared to free MEL. pacMEL also significantly reduces hepatic damage and unwanted innate immune response and all but eliminated hemolytic activities of MEL. Importantly, pacMEL passively accumulates at subcutaneously inoculated tumor sites and exhibits stronger tumor-suppressive activity than molecular MEL. Collectively, pacMEL makes MEL a safer and more appealing drug candidate.


Subject(s)
Antineoplastic Agents/therapeutic use , Melitten/analogs & derivatives , Melitten/therapeutic use , Neoplasms/drug therapy , Polyethylene Glycols/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Cell Line, Tumor , Female , Humans , Melitten/pharmacokinetics , Melitten/toxicity , Mice, Inbred C57BL , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/toxicity , Xenograft Model Antitumor Assays
8.
Biochemistry ; 49(36): 7920-9, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20695504

ABSTRACT

Melittin is a good model antimicrobial peptide to understand the basis of its lytic activities against bacteria and mammalian cells. Novel analogues of melittin were designed by substituting the leucine residue(s) at the "d" and "a" positions of its previously identified leucine zipper motif. A scrambled peptide having the same composition of melittin with altered leucine zipper sequence was also designed. The analogues of melittin including the scrambled peptide showed a drastic reduction in cytotoxicity though they exhibited comparable bactericidal activities. Only melittin but not its analogues localized strongly onto hRBCs and formed pores of approximately 2.2-3.4 nm. However, melittin and its analogues localized similarly onto Escherichia coli and formed pores of varying sizes as tested onto Bacillus megaterium. The data showed that the substitution of hydrophobic leucine residue(s) by lesser hydrophobic alanine residue(s) in the leucine zipper sequence of melittin disturbed its pore-forming activity and mechanism only in hRBCs but not in the tested bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Erythrocytes/drug effects , Escherichia coli/drug effects , Melitten/analogs & derivatives , Melitten/pharmacology , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Erythrocytes/metabolism , Escherichia coli/metabolism , Hemolysis , Humans , Hydrophobic and Hydrophilic Interactions , Leucine Zippers , Microscopy, Confocal , Molecular Sequence Data
9.
Endocr Relat Cancer ; 16(2): 549-64, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19261682

ABSTRACT

Lytic peptide Hecate (23-amino acid (AA)) fused with a 15-AA fragment of human chorionic gonadotropin-beta (CG-beta), Hecate-CGbeta conjugate (H-CGbeta-c) selectively binds to and destroys tumor cells expressing LH/chorionic gonadotropin receptor (Lhcgr). Transgenic mice (6.5 month old) expressing SV40 T-antigen under the inhibin-alpha promoter (inhalpha/Tag) presenting with Lhcgr expressing adrenal tumors were treated either with H-CGbeta-c, GnRH antagonist (GnRH-a), estradiol (E(2); only females) or their combinations for 1 month. We expected that GnRH-a or E(2) in combination with H-CGbeta-c could improve the treatment efficacy especially in females by decreasing circulating LH and eliminating the potential competition of serum LH with the H-CGbeta-c. GnRH-a and H-CGbeta-c treatments were successful in males (adrenal weights 14 +/- 2.8 mg and 60 +/- 26 vs 237 +/- 59 mg in controls; P < 0.05). Histopathologically, GnRH-a apparently destroyed the adrenal parenchyma leaving only the fibrotic capsule with few necrotic foci. In females, H-CGbeta-c was totally ineffective, whereas GnRH-a (19 +/- 5 mg) or E(2) (77 +/- 50 mg) significantly reduced the adrenal weights compared with controls (330 +/- 70 mg). Adrenal morphometry, cell proliferation markers, post-treatment suppression of serum progesterone, and quantitative RT-PCR of GATA-4, Lhcgr, and GATA-6 further supported the positive outcome. H-CGbeta-c selectively killed the Lhcgr expressing tumor cells, whereas GnRH-a blocked tumor progression through gonadotropin suppression, emphasizing the gonadotropin dependency of these adrenocortical tumors. If extrapolated to humans, H-CGbeta-c could be considered for the treatment of gonadotropin-dependent adrenal tumors in males, whereas in females gonadotropin suppression, but not H-CGbeta-c, would work better.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Antigens, Viral, Tumor/metabolism , Gonadotropins/metabolism , Inhibins/genetics , Melitten/analogs & derivatives , Promoter Regions, Genetic/genetics , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Animals , Cell Proliferation , Chorionic Gonadotropin, beta Subunit, Human/therapeutic use , Female , Gonadotropin-Releasing Hormone/metabolism , Humans , Immunoenzyme Techniques , Male , Melitten/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Transgenic , Progesterone/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Simian virus 40/genetics
10.
Endocr Relat Cancer ; 15(2): 635-48, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18509010

ABSTRACT

Novel strategies are needed for the treatment of adrenocortical tumors that are usually resistant to chemotherapy. Hecate, a 23-amino acid lytic peptide, was conjugated to the 15-amino acid (81-95) fragment of the human chorionic gonadotropin beta (CGbeta) chain, which would selectively kill cancer cells expressing the LH receptor (LHR) sparing the normal ones with LHR. To prove the principle that Hecate-CGbeta conjugate may eradicate tumors ectopically expressing plasma membrane receptors, transgenic (TG) inhibin alpha-subunit promoter (inhalpha)/Simian Virus 40 T-antigen mice, expressing LHR in their adrenal gland tumors, were used as the experimental model. Wild-type control littermates and TG mice with adrenal tumors were treated with either Hecate or Hecate-CGbeta conjugate at the age of 6.5 months for 3 weeks and killed 7 days after the last treatment. The Hecate-CGbeta conjugate reduced the adrenal tumor burden significantly in TG male but not in female mice, in comparison with Hecate-treated mice. Hecate-CGbeta conjugate treatment did not affect normal adrenocortical function as the serum corticosterone level between Hecate and Hecate-CGbeta conjugate groups were similar. The mRNA and protein expressions of GATA-4 and LHR colocalized only in tumor area, and a significant downregulation of gene expression was found after the Hecate-CGbeta conjugate in comparison with Hecate- and/or non-treated adrenal tumors by western blotting. This finding provides evidence for a selective destruction of the tumor cells by the Hecate-CGbeta conjugate. Hereby, our findings support the principle that Hecate-CGbeta conjugate is able to specifically destroy tumor cells that ectopically express LHR.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Adrenocortical Adenoma/drug therapy , Antineoplastic Agents/pharmacology , Melitten/analogs & derivatives , Receptors, LH/genetics , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Adrenocortical Adenoma/genetics , Adrenocortical Adenoma/pathology , Animals , Chorionic Gonadotropin, beta Subunit, Human/pharmacology , Disease Models, Animal , Female , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Luteinizing Hormone/blood , Male , Melitten/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Receptors, LH/metabolism
11.
Reprod Biol ; 8(2): 135-47, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18677401

ABSTRACT

Hecate-betaCG and Phor14-betaCG(ala) are relatively short, amphipathic alpha-helical cationic peptides with the ability to destroy selectively breast, prostate and ovarian cancer cells. Treatment with proteins and peptides frequently initiated antibody formation. Short peptides may minimize the risk of the immune system mobilization after treatment but it is necessary to investigate whether Hecate-betaCG and Phor14-betaCG(ala) induce the immune system to produce antibody and whether they affect the reproductive organs in normal wild-type mice. The results of our experiments showed that specific antibodies, tested by the enzyme-immunoassay, were not detected in the group treated with Hecate-betaCG and Phor14-betaCG(ala). The blood concentrations of both peptides begun to decrease from 60 minutes after injection and after 240 minutes its levels were undetectable. Histopatho-logical examination exhibited degenerative changes in the prostate glands and testes in males and in the ovaries and uteri of females treated with both peptides. In conclusion, our results indicate that both relatively small and rapidly metabolized peptides are not immunogenic and can be used for further investigation as a potential cancer treatment.


Subject(s)
Chorionic Gonadotropin/immunology , Melitten/analogs & derivatives , Peptide Fragments/immunology , Animals , Antibody Formation , Chorionic Gonadotropin, beta Subunit, Human/immunology , Female , Goats/immunology , Male , Melitten/immunology , Mice , Mice, Inbred BALB C/immunology
12.
Int J Biochem Cell Biol ; 101: 39-48, 2018 08.
Article in English | MEDLINE | ID: mdl-29800725

ABSTRACT

Chemotherapy resistance represents a major obstacle in the treatment of patients with hepatocellular carcinoma (HCC). The purpose of this study was to investigate the anti-cancer effect of MEL-pep, a novel analog of the natural antibacterial peptide melittin (MEL), on human 5-fluorouracil-resistant HCC cells (BEL-7402/5-FU) and to clarify the molecular mechanisms involved in these effects. We found that MEL-pep inhibited the proliferation of BEL-7402/5-FU cells and reversed 5-FU resistance in vitro. MEL-pep directly bound to BEL-7402/5-FU cells and disrupted the cell membrane. P-glycoprotein (P-gp) plays an important role in the development of resistance to anticancer drugs. We found that MEL-pep inhibited P-gp expression and increased the intracellular accumulation of the P-gp substrate rhodamine-123 in BEL-7402/5-FU cells. Additionally, the phosphorylation of Akt and NF-κB/p65 nuclear translocation was all inhibited by MEL-pep. Insulin - like growth factor I, a phosphatidylinositol 3 kinase(PI3K) /protein kinase B(AKT) agonist, reversed MEL-pep induced P-gp suppression. Therefore, MEL-pep inhibited P-gp expression by deactivating the PI3K/Akt signaling pathway. Finally, in a BEL-7402/5-FU cell-derived xenograft tumor model in mice, we found that the intratumoral administration of MEL-pep inhibited tumor growth in a dose-dependent manner. Thus, MEL-pep could be a promising candidate in the treatment of chemotherapy resistant HCC.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/drug therapy , Melitten/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Inhibitory Concentration 50 , Insulin-Like Growth Factor I/pharmacology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Melitten/analogs & derivatives , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Mol Cell Endocrinol ; 269(1-2): 17-25, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17363137

ABSTRACT

Improvement of cancer treatment is a major challenge of medical research. Despite the immense efforts made in the improvement of diagnosis and treatment, cancer remains a major concern and cause of morbidity and mortality. Most of the modern anti-neoplastic therapies have severe side effects, and tumor cells often develop drug resistance. There is promise in the new generation of treatments (gene therapy, immunotherapy, vaccines, etc.) that are under development, but the efficacies and side effects of such therapies have so far been disappointing. Receptor-based therapies are not new, but many normal cells also present the same receptors reducing the specificity of such approaches. Several lytic peptides have been investigated because of they appear to kill cancer cells due to changes of their membrane potential. Thus, linking receptor-specific ligands to lytic peptides is expected to augment the specificity of targeting and decrease the toxicity of lytic peptides on normal cells. One such polypeptide is hecate (an analogue to the bee venom main component, melittin) that preferentially kills cancer cells at low doses. When this peptide is fused with the 81-95 amino acid fragment of chorionic gonadotropin-beta (CGbeta) subunit (hecate-CGbeta), it targets cells expressing luteinizing hormone receptor (LHR), even at very low doses, or when LHR is expressed at low level. Our recent data showed that this peptide conjugate is efficient in destroying LHR-positive cells in xenografts and more importantly in transgenic mouse models developing LHR-positive somatic cell tumors in gonads. The mechanism of action of hecate-CGbeta after binding to LHR is destruction of cell membranes resulting in rapid cell death by necrosis with minimal side effects. This review summarizes our findings on the action of this novel peptide and considers the future potential of this family of targeting peptides in the treatment of neoplasias.


Subject(s)
Melitten/analogs & derivatives , Ovarian Neoplasms/drug therapy , Receptors, LH/metabolism , Testicular Neoplasms/drug therapy , Animals , Chorionic Gonadotropin, beta Subunit, Human/therapeutic use , Drug Delivery Systems , Female , Genetic Therapy , Humans , Male , Melitten/therapeutic use , Models, Biological , Ovarian Neoplasms/metabolism , Receptors, LH/antagonists & inhibitors , Receptors, LH/genetics , Testicular Neoplasms/metabolism
14.
Mol Cell Endocrinol ; 269(1-2): 26-33, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17382461

ABSTRACT

In a series of in vivo and in vitro experiments, it was shown that membrane disrupting lytic peptides (Hecate, Phor14, or Phor21) conjugated to a 15 amino acid segment of the beta chain of CG or to LHRH were able to target and destroy hormone dependent and independent human prostate cancer xenografts in nude mice. In vitro sensitivity of the cells to the drugs was directly related to LH/CG receptor expression, and pretreatment in vitro or in vivo with estrogens or FSH to enhance LH/CG receptor expression capacity and increased sensitivity to the drugs. Administration of unconjugated Hecate and LHRH was ineffective. Most importantly, all of the lytic peptide-betaCG conjugates tested were highly effective in destroying prostate cancer metastatic cells in lymph nodes, bones and lungs.


Subject(s)
Carcinoma/drug therapy , Gonadotropin-Releasing Hormone/therapeutic use , Melitten/analogs & derivatives , Neoplasm Metastasis/drug therapy , Prostatic Neoplasms/drug therapy , Animals , Carcinoma/pathology , Cell Survival/drug effects , Chorionic Gonadotropin, beta Subunit, Human/pharmacology , Chorionic Gonadotropin, beta Subunit, Human/therapeutic use , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/pharmacology , Humans , Male , Melitten/pharmacology , Melitten/therapeutic use , Necrosis/chemically induced , Prostatic Neoplasms/pathology
15.
Mol Cell Endocrinol ; 260-262: 183-9, 2007 Jan 02.
Article in English | MEDLINE | ID: mdl-17101210

ABSTRACT

In a series of in vivo and in vitro experiments, the concept has been established that breast cancer cells that express LH/CG or LHRH receptors can be targeted and destroyed by constructs consisting of a lytic peptide moiety and a 15-amino acid segment of the beta-chain of CG or by an LHRH lytic peptide conjugate. Data obtained in vitro established the validity of this concept, showed the specificities of the Hecate-betaCG, and Phor14 and Phor21-betaCG conjugates in killing cells that express functional LH/CG receptors and proved that the LH/CG receptor capacity is directly related to the compound's specificity. In in vivo experiments, Hecate-betaCG, Phor14-betaCG, and Phor21-betaCG(ala) each caused highly significant reductions of tumor volume and tumor burden in nude mice bearing breast cancer xenografts; Hecate and Phor21 alone or conjugated with non-specific peptides were not effective. Most importantly, the lytic peptide conjugates were all highly effective in targeting and destroying disseminated breast cancer metastases in lymph nodes, bones, lungs and other organs.


Subject(s)
Breast Neoplasms/drug therapy , Chorionic Gonadotropin, beta Subunit, Human/therapeutic use , Gonadotropin-Releasing Hormone/therapeutic use , Melitten/analogs & derivatives , Peptide Fragments/therapeutic use , Peptides/therapeutic use , Amino Acid Sequence , Animals , Cell Death , Chorionic Gonadotropin, beta Subunit, Human/chemistry , Female , Luciferases/metabolism , Lung/pathology , Lymph Nodes/pathology , Melitten/chemistry , Melitten/therapeutic use , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Transplantation , Peptide Fragments/chemistry , Peptides/chemistry , Spinal Cord/pathology , Tumor Burden
16.
J Biochem Mol Biol ; 40(6): 1090-4, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-18047808

ABSTRACT

Melittin (ME), a linear 26-residue non-cell-selective antimicrobial peptide, displays strong lytic activity against bacterial and human red blood cells. To design ME analogue with improved cell selectivity, we synthesized a melittin diastereomer (ME-D) with D-amino acid in the leucine zipper sequence (Leu-6, Lue-13 and Ile-20). Compared to ME, ME-D exhibited the same or 2-fold higher antibacterial activity but 8-fold less hemolytic activity. Circular dichroism analysis revealed that ME-D has much less alpha-helical content in alpha-helical content in the presence of zwitterionic EYPC/cholesterol (10 : 1, w/w) liposomes compared to negatively charged EYPE/EYPG (7 : 3, w/w) liposomes. The blue shift of the fluorescence emission maximum of ME-D in zwitterionic EYPC/ cholesterol (10 : 1, w/w) liposomes was much smaller than in negatively charged EYPE/EYPG (7 : 3, w/w) liposomes. These results suggested that the improvement in therapeutic index/cell selectivity of ME-D is correlated with its less permeability to zwitterionic membranes.


Subject(s)
Melitten/analogs & derivatives , Amino Acid Sequence , Bacteria/drug effects , Circular Dichroism , Drug Design , Hemolysis/drug effects , Humans , In Vitro Techniques , Leucine Zippers/genetics , Melitten/chemistry , Melitten/genetics , Melitten/pharmacology , Molecular Sequence Data , Protein Structure, Secondary , Stereoisomerism
17.
Nat Biotechnol ; 18(2): 225-7, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10657134

ABSTRACT

The increased resistance of various bacteria toward available antibiotic drugs has initiated intensive research efforts into identifying new sources of antimicrobial substances. Short antibiotic peptides (10-30 residues) are prevalent in nature as part of the intrinsic defense mechanisms of most organisms and have been proposed as a blueprint for the design of novel antimicrobial agents. Antimicrobial peptides are generally believed to kill bacteria through membrane permeabilization and extensive pore-formation. Assays providing rapid and easy evaluation of interactions between antimicrobial membrane peptides and lipid bilayers could significantly improve screening for substances with effective antibacterial properties, as well as contribute to the elucidation of structural and functional properties of antimicrobial peptides. Here we describe a colorimetric sensor in which particles composed of phospholipids and polymerized polydiacetylene (PDA) lipids were shown to exhibit striking color changes upon interactions with antimicrobial membrane peptides. The color changes in the system occur because of the structural perturbation of the lipids following their interactions with antimicrobial peptides. The assay was also sensitive to the antibacterial properties of structurally and functionally related peptide analogs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Colorimetry/methods , Microbial Sensitivity Tests/methods , Peptides/pharmacology , Acetylene/analogs & derivatives , Alamethicin/analogs & derivatives , Alamethicin/pharmacology , Melitten/analogs & derivatives , Melitten/pharmacology , Microsomes/drug effects , Oligopeptides/pharmacology , Permeability , Phospholipids , Polyacetylene Polymer , Polymers , Polyynes , Transducers
18.
Cancer Lett ; 402: 16-31, 2017 08 28.
Article in English | MEDLINE | ID: mdl-28536009

ABSTRACT

Melittin (MEL), a major peptide component of bee venom, is an attractive candidate for cancer therapy. This agent has shown a variety of anti-cancer effects in preclinical cell culture and animal model systems. Despite a convincing efficacy data against variety of cancers, its applicability to humans has met with challenges due to several issues including its non-specific cytotoxicity, degradation and hemolytic activity. Several optimization approaches including utilization of nanoparticle based delivery of MEL have been utilized to circumvent the issues. Here, we summarize the current understanding of the anticancer effects of bee venom and MEL on different kinds of cancers. Further, we also present the available information for the possible mechanism of action of bee venom and/or MEL.


Subject(s)
Antineoplastic Agents/therapeutic use , Melitten/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Drug Carriers , Drug Compounding , Drug Stability , Humans , Melitten/adverse effects , Melitten/analogs & derivatives , Melitten/chemistry , Nanoparticles , Nanotechnology/methods , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/drug effects
19.
J Zhejiang Univ Sci B ; 18(6): 522-531, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28585428

ABSTRACT

Hepatocarcinoma is one of the malignant cancers with significant morbidity and mortality. Immunotherapy has emerged in clinical treatment, owing to the limitation and severe side effects of chemotherapy. In the immune system, natural killer (NK) cells are important effectors required to eliminate malignant tumor cells without the limitation of major histocompatibility complex (MHC) molecule issues. Hence, treatment which could stimulate NK cells is of great interest. Here, we investigated the efficacy of the combined therapy of TT-1 (a mutant of melittin) and interferon-α (IFN-α) on NK cells and human liver cancer HepG-2/Huh7 cells in vitro and in vivo, as well as the mechanism involved. The combination therapy significantly inhibited the growth of HepG-2/Huh7 cells in vivo, but this effect was impaired after depleting NK cells. TT-1 not only up-regulated MHC class I-related chain molecules A (MICA) expression, but also prevented the secretion of soluble MICA (sMICA). Both the mRNA and protein of a disintegrin and metallopeptidase 10 (ADAM 10) in HepG-2/Huh7 cells were decreased after TT-1 treatment. The combined therapy of TT-1 and IFN-α could suppress the growth of HepG-2/Huh7 xenografted tumor effectively via promoting the interaction of NK group 2, member D (NKG2D) and MICA, indicating that TT-1+IFN-α would be a potential approach in treating liver cancer.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Histocompatibility Antigens Class I/metabolism , Interferon-alpha/administration & dosage , Liver Neoplasms/drug therapy , Melitten/analogs & derivatives , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Adjuvants, Immunologic/administration & dosage , Amino Acid Sequence , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Hep G2 Cells , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Melitten/administration & dosage , Melitten/genetics , Mice , Mice, Nude , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Xenograft Model Antitumor Assays
20.
J Control Release ; 112(2): 240-8, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16545884

ABSTRACT

Melittin-polyethylenimine (PEI) conjugates have been shown to enhance gene transfer efficiency of polyplexes due to their membrane-destabilizing properties. Inherent lytic activity at neutral pH however also provokes high cytotoxicity due to plasma membrane damage. In order to shift the lytic activity towards the endosomal membrane, several melittin analogs were designed. Acidic modification of melittin by replacing neutral glutamines (Gln-25 and Gln-26) with glutamic acid residues greatly improved the lytic activity of C-terminally linked PEI conjugates at the endosomal pH of 5. This activity correlated well with the gene transfer efficiency of polyplexes in four different cell lines. Melittin-PEI conjugates with high lytic activities at endosomal pH were then incorporated into EGF receptor-targeted and polyethylene glycol-shielded polyplexes. The resulting particles had virus-like dimension (150 nm) with a neutral surface charge and were subsequently purified by size exclusion chromatography to remove unbound toxic PEI conjugate. These purified polyplexes mediated EGF-receptor-specific gene transfer with up to 70-fold higher activity compared to the corresponding PEI polyplexes without melittin.


Subject(s)
DNA/administration & dosage , Endosomes/chemistry , Melitten/analogs & derivatives , Melitten/administration & dosage , Polyethyleneimine/administration & dosage , Transfection , Animals , Cell Line , Erythrocytes/cytology , Erythrocytes/drug effects , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , Hydrogen-Ion Concentration , Luciferases/genetics , Melitten/adverse effects , Mice , Particle Size , Polyethyleneimine/adverse effects , Polyethyleneimine/chemistry , Rats
SELECTION OF CITATIONS
SEARCH DETAIL