Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 21(1): 30, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36737758

ABSTRACT

BACKGROUND: C3G is a guanine nucleotide exchange factor (GEF) that activates Rap1 to promote cell adhesion. Resting C3G is autoinhibited and the GEF activity is released by stimuli that signal through tyrosine kinases. C3G is activated by tyrosine phosphorylation and interaction with Crk adaptor proteins, whose expression is elevated in multiple human cancers. However, the molecular details of C3G activation and the interplay between phosphorylation and Crk interaction are poorly understood. METHODS: We combined biochemical, biophysical, and cell biology approaches to elucidate the mechanisms of C3G activation. Binding of Crk adaptor proteins to four proline-rich motifs (P1 to P4) in C3G was characterized in vitro using isothermal titration calorimetry and sedimentation velocity, and in Jurkat and HEK293T cells by affinity pull-down assays. The nucleotide exchange activity of C3G over Rap1 was measured using nucleotide-dissociation kinetic assays. Jurkat cells were also used to analyze C3G translocation to the plasma membrane and the C3G-dependent activation of Rap1 upon ligation of T cell receptors. RESULTS: CrkL interacts through its SH3N domain with sites P1 and P2 of inactive C3G in vitro and in Jurkat and HEK293T cells, and these sites are necessary to recruit C3G to the plasma membrane. However, direct stimulation of the GEF activity requires binding of Crk proteins to the P3 and P4 sites. P3 is occluded in resting C3G and is essential for activation, while P4 contributes secondarily towards complete stimulation. Tyrosine phosphorylation of C3G alone causes marginal activation. Instead, phosphorylation primes C3G lowering the concentration of Crk proteins required for activation and increasing the maximum activity. Unexpectedly, optimal activation also requires the interaction of CrkL-SH2 domain with phosphorylated C3G. CONCLUSION: Our study revealed that phosphorylation of C3G by Src and Crk-binding form a two-factor mechanism that ensures tight control of C3G activation. Additionally, the simultaneous SH2 and SH3N interaction of CrkL with C3G, required for the activation, reveals a novel adaptor-independent function of Crk proteins relevant to understanding their role in physiological signaling and their deregulation in diseases. Video abstract.


Subject(s)
Guanine Nucleotide-Releasing Factor 2 , Nuclear Proteins , Humans , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide-Releasing Factor 2/metabolism , HEK293 Cells , Nuclear Proteins/metabolism , Nucleotides/metabolism , Proto-Oncogene Proteins c-crk/metabolism , src Homology Domains , Tyrosine/metabolism
2.
Cancer Res ; 81(21): 5438-5450, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34417202

ABSTRACT

Despite the approval of several multikinase inhibitors that target SRC and the overwhelming evidence of the role of SRC in the progression and resistance mechanisms of many solid malignancies, inhibition of its kinase activity has thus far failed to improve patient outcomes. Here we report the small molecule eCF506 locks SRC in its native inactive conformation, thereby inhibiting both enzymatic and scaffolding functions that prevent phosphorylation and complex formation with its partner FAK. This mechanism of action resulted in highly potent and selective pathway inhibition in culture and in vivo. Treatment with eCF506 resulted in increased antitumor efficacy and tolerability in syngeneic murine cancer models, demonstrating significant therapeutic advantages over existing SRC/ABL inhibitors. Therefore, this mode of inhibiting SRC could lead to improved treatment of SRC-associated disorders. SIGNIFICANCE: Small molecule-mediated inhibition of SRC impairing both catalytic and scaffolding functions confers increased anticancer properties and tolerability compared with other SRC/ABL inhibitors.


Subject(s)
Bone Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Focal Adhesion Kinase 1/antagonists & inhibitors , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Apoptosis , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Conformation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , src-Family Kinases/chemistry , src-Family Kinases/metabolism
3.
Front Mol Biosci ; 8: 648468, 2021.
Article in English | MEDLINE | ID: mdl-33816561

ABSTRACT

Bacterial conjugation is the main horizontal gene transfer route responsible for the spread of antibiotic resistance, virulence and toxin genes. During conjugation, DNA is transferred from a donor to a recipient cell via a sophisticated channel connecting the two cells. Conjugation not only affects many different aspects of the plasmid and the host, ranging from the properties of the membrane and the cell surface of the donor, to other developmental processes such as competence, it probably also poses a burden on the donor cell due to the expression of the large number of genes involved in the conjugation process. Therefore, expression of the conjugation genes must be strictly controlled. Over the past decade, the regulation of the conjugation genes present on the conjugative Bacillus subtilis plasmid pLS20 has been studied using a variety of methods including genetic, biochemical, biophysical and structural approaches. This review focuses on the interplay between RcopLS20, RappLS20 and Phr*pLS20, the proteins that control the activity of the main conjugation promoter P c located upstream of the conjugation operon. Proper expression of the conjugation genes requires the following two fundamental elements. First, conjugation is repressed by default and an intercellular quorum-signaling system is used to sense conditions favorable for conjugation. Second, different layers of regulation act together to repress the P c promoter in a strict manner but allowing rapid activation. During conjugation, ssDNA is exported from the cell by a membrane-embedded DNA translocation machine. Another membrane-embedded DNA translocation machine imports ssDNA in competent cells. Evidences are reviewed indicating that conjugation and competence are probably mutually exclusive processes. Some of the questions that remain unanswered are discussed.

4.
Nucleic Acids Res ; 48(19): 10785-10801, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33045732

ABSTRACT

Quorum sensing plays crucial roles in bacterial communication including in the process of conjugation, which has large economical and health-related impacts by spreading antibiotic resistance. The conjugative Bacillus subtilis plasmid pLS20 uses quorum sensing to determine when to activate the conjugation genes. The main conjugation promoter, Pc, is by default repressed by a regulator RcopLS20 involving DNA looping. A plasmid-encoded signalling peptide, Phr*pLS20, inactivates the anti-repressor of RcopLS20, named RappLS20, which belongs to the large group of RRNPP family of regulatory proteins. Here we show that DNA looping occurs through interactions between two RcopLS20 tetramers, each bound to an operator site. We determined the relative promoter strengths for all the promoters involved in synthesizing the regulatory proteins of the conjugation genes, and constructed an in vivo system uncoupling these regulatory genes to show that RappLS20 is sufficient for activating conjugation in vivo. We also show that RappLS20 actively detaches RcopLS20 from DNA by preferentially acting on the RcopLS20 molecules involved in DNA looping, resulting in sequestration but not inactivation of RcopLS20. Finally, results presented here in combination with our previous results show that activation of conjugation inhibits competence and competence development inhibits conjugation, indicating that both processes are mutually exclusive.


Subject(s)
Bacillus subtilis/genetics , Bacterial Proteins/genetics , Conjugation, Genetic , Gene Expression Regulation, Bacterial , Quorum Sensing , Bacillus subtilis/metabolism , Bacillus subtilis/physiology , Bacterial Proteins/metabolism , Plasmids/genetics , Promoter Regions, Genetic
5.
Nucleic Acids Res ; 48(14): 8113-8127, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32658272

ABSTRACT

Quorum sensing allows bacterial cells to communicate through the release of soluble signaling molecules into the surrounding medium. It plays a pivotal role in controlling bacterial conjugation in Gram-positive cells, a process that has tremendous impact on health. Intracellular regulatory proteins of the RRNPP family are common targets of these signaling molecules. The RRNPP family of gene regulators bind signaling molecules at their C-terminal domain (CTD), but have highly divergent functionalities at their N-terminal effector domains (NTD). This divergence is also reflected in the functional states of the proteins, and is highly interesting from an evolutionary perspective. RappLS20 is an RRNPP encoded on the Bacillus subtilis plasmid pLS20. It relieves the gene repression effectuated by RcopLS20 in the absence of the mature pLS20 signaling peptide Phr*pLS20. We report here an in-depth structural study of apo and Phr*pLS20-bound states of RappLS20 at various levels of atomic detail. We show that apo-RappLS20 is dimeric and that Phr*pLS20-bound Rap forms NTD-mediated tetramers. In addition, we show that RappLS20 binds RcopLS20 directly in the absence of Phr*pLS20 and that addition of Phr*pLS20 releases RcopLS20 from RappLS20. This allows RcopLS20 to bind the promotor region of crucial conjugation genes blocking their expression.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Operon , Protein Multimerization , Trans-Activators/metabolism , Bacillus subtilis , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Conjugation, Genetic/genetics , Peptides/metabolism , Promoter Regions, Genetic , Tetratricopeptide Repeat , Trans-Activators/chemistry , Trans-Activators/genetics
6.
Nucleic Acids Res ; 46(22): 11910-11926, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30380104

ABSTRACT

The principal route for dissemination of antibiotic resistance genes is conjugation by which a conjugative DNA element is transferred from a donor to a recipient cell. Conjugative elements contain genes that are important for their establishment in the new host, for instance by counteracting the host defense mechanisms acting against incoming foreign DNA. Little is known about these establishment genes and how they are regulated. Here, we deciphered the regulation mechanism of possible establishment genes of plasmid p576 from the Gram-positive bacterium Bacillus pumilus. Unlike the ssDNA promoters described for some conjugative plasmids, the four promoters of these p576 genes are repressed by a repressor protein, which we named Reg576. Reg576 also regulates its own expression. After transfer of the DNA, these genes are de-repressed for a period of time until sufficient Reg576 is synthesized to repress the promoters again. Complementary in vivo and in vitro analyses showed that different operator configurations in the promoter regions of these genes lead to different responses to Reg576. Each operator is bound with extreme cooperativity by two Reg576-dimers. The X-ray structure revealed that Reg576 has a Ribbon-Helix-Helix core and provided important insights into the high cooperativity of DNA recognition.


Subject(s)
Bacillus pumilus/genetics , Bacterial Proteins/chemistry , DNA/chemistry , Gene Transfer, Horizontal , Plasmids/chemistry , Repressor Proteins/chemistry , Bacillus pumilus/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Binding Sites , Cloning, Molecular , Conjugation, Genetic , DNA/genetics , DNA/metabolism , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gene Expression Regulation, Bacterial , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Nucleic Acid Conformation , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Shigella flexneri/genetics , Shigella flexneri/metabolism
7.
Front Microbiol ; 8: 2138, 2017.
Article in English | MEDLINE | ID: mdl-29163424

ABSTRACT

Bacterial conjugation is the process by which a conjugative element (CE) is transferred horizontally from a donor to a recipient cell via a connecting pore. One of the first steps in the conjugation process is the formation of a nucleoprotein complex at the origin of transfer (oriT), where one of the components of the nucleoprotein complex, the relaxase, introduces a site- and strand specific nick to initiate the transfer of a single DNA strand into the recipient cell. In most cases, the nucleoprotein complex involves, besides the relaxase, one or more additional proteins, named auxiliary proteins, which are encoded by the CE and/or the host. The conjugative plasmid pLS20 replicates in the Gram-positive Firmicute bacterium Bacillus subtilis. We have recently identified the relaxase gene and the oriT of pLS20, which are separated by a region of almost 1 kb. Here we show that this region contains two auxiliary genes that we name aux1LS20 and aux2LS20 , and which we show are essential for conjugation. Both Aux1LS20 and Aux2LS20 are predicted to contain a Ribbon-Helix-Helix DNA binding motif near their N-terminus. Analyses of the purified proteins show that Aux1LS20 and Aux2LS20 form tetramers and hexamers in solution, respectively, and that they both bind preferentially to oriTLS20 , although with different characteristics and specificities. In silico analyses revealed that genes encoding homologs of Aux1LS20 and/or Aux2LS20 are located upstream of almost 400 relaxase genes of the RelLS20 family (MOBL) of relaxases. Thus, Aux1LS20 and Aux2LS20 of pLS20 constitute the founding member of the first two families of auxiliary proteins described for CEs of Gram-positive origin.

8.
Structure ; 25(7): 1145-1152.e4, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28648606

ABSTRACT

The R2TP complex, comprising the Rvb1p-Rvb2p AAA-ATPases, Tah1p, and Pih1p in yeast, is a specialized Hsp90 co-chaperone required for the assembly and maturation of multi-subunit complexes. These include the small nucleolar ribonucleoproteins, RNA polymerase II, and complexes containing phosphatidylinositol-3-kinase-like kinases. The structure and stoichiometry of yeast R2TP and how it couples to Hsp90 are currently unknown. Here, we determine the 3D organization of yeast R2TP using sedimentation velocity analysis and cryo-electron microscopy. The 359-kDa complex comprises one Rvb1p/Rvb2p hetero-hexamer with domains II (DIIs) forming an open basket that accommodates a single copy of Tah1p-Pih1p. Tah1p-Pih1p binding to multiple DII domains regulates Rvb1p/Rvb2p ATPase activity. Using domain dissection and cross-linking mass spectrometry, we identified a unique region of Pih1p that is essential for interaction with Rvb1p/Rvb2p. These data provide a structural basis for understanding how R2TP couples an Hsp90 dimer to a diverse set of client proteins and complexes.


Subject(s)
Adenosine Triphosphatases/chemistry , HSP90 Heat-Shock Proteins/chemistry , Molecular Chaperones/chemistry , Nuclear Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Transcription Factors/chemistry , Adenosine Triphosphatases/metabolism , Binding Sites , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Protein Binding , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism
9.
Biochemistry ; 53(10): 1548-50, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24564530

ABSTRACT

ß-Lactam antibiotics have faced obsolescence with the emergence of methicillin-resistant Staphylococcus aureus (MRSA). A complex set of events ensues upon exposure of MRSA to these antibiotics, which culminates in proteolysis of BlaI or MecI, two gene repressors, and results in the induction of resistance. We report studies on the mechanism of binding of these gene repressors to the operator regions by fluorescence anisotropy. Within the range of in vivo concentrations for BlaI and MecI, these proteins interact with their regulatory elements in a reversible manner, as both a monomer and a dimer.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Methicillin-Resistant Staphylococcus aureus/metabolism , Repressor Proteins/metabolism , beta-Lactams/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Drug Resistance, Bacterial , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Operator Regions, Genetic , Operon , Promoter Regions, Genetic , Repressor Proteins/chemistry , Repressor Proteins/genetics
10.
Amino Acids ; 46(4): 1047-58, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24445871

ABSTRACT

Therapeutic application of many drugs is often hampered by poor or denied access to intracellular targets. A case in point is miltefosine (MT), an orally active antiparasitic drug, which becomes ineffective when parasites develop dysfunctional uptake systems. We report here the synthesis of a fluorescent BODIPY-embedding MT analogue with appropriate thiol functionalization allowing linkage to the cell-penetrating Tat(48-60) peptide through disulfide or thioether linkages. The resulting constructs are efficiently internalized into the otherwise MT-invulnerable R40 Leishmania strain, resulting in fast parasite killing, and hence successful avoidance of the resistance. In the disulfide-linked conjugate, an additional fluoro tag on the Tat moiety allows to monitor its reductive cleavage within the cytoplasm. Terminally differentiated cells such as peritoneal macrophages, impervious to MT unless infected by Leishmania, can uptake the drug in its Tat-conjugated form. The results afford proof-of-principle for using CPP vectors to avert drug resistance in parasites, and/or for tackling leishmaniasis by modulating macrophage uptake.


Subject(s)
Anthelmintics/chemical synthesis , Anthelmintics/pharmacokinetics , Boron Compounds/chemistry , Boron Compounds/pharmacokinetics , Cell-Penetrating Peptides/chemistry , Leishmania/drug effects , Phosphorylcholine/analogs & derivatives , tat Gene Products, Human Immunodeficiency Virus/chemistry , Anthelmintics/chemistry , Cell Line , Cell-Penetrating Peptides/pharmacokinetics , Drug Design , Humans , Leishmaniasis/drug therapy , Leishmaniasis/parasitology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/parasitology , Molecular Structure , Phosphorylcholine/chemistry , tat Gene Products, Human Immunodeficiency Virus/pharmacokinetics
11.
Mol Pharm ; 7(5): 1608-17, 2010 Oct 04.
Article in English | MEDLINE | ID: mdl-20715776

ABSTRACT

IB-01212, an antitumoral cyclodepsipeptide isolated from the mycelium of the marine fungus Clonostachys sp., showed leishmanicidal activity at a low micromolar range of concentrations on promastigote and amastigote forms of the parasite. Despite its cationic and amphipathic character, shared with other membrane active antibiotic peptides, IB-01212 did not cause plasma membrane lesions large enough to allow the entrance of the vital dye SYTOX green (MW = 600), even at concentrations causing full lethality of the parasite. Having ruled out massive disruption of the plasma membrane, we surmised the involvement of intracellular targets. Proof of concept for this assumption was provided by the mitochondrial dysfunction caused by IB-01212, which finally caused the death of the parasite through an apoptotic-like process. The size of the cycle, the preservation of the C2 symmetry, and the nature of the bonds linking the two tetrapeptide halves participate in the modulation of the leishmanicidal activity exerted by this compound. Here we discuss the potential of IB-01212 as a lead for new generations of surrogates to be used in chemotherapy treatments against Leishmania .


Subject(s)
Antiprotozoal Agents/pharmacology , Depsipeptides/pharmacology , Leishmania/drug effects , Animals , Antineoplastic Agents/pharmacology , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/chemistry , Apoptosis/drug effects , Cell Membrane/drug effects , Depsipeptides/administration & dosage , Depsipeptides/chemistry , Fungal Proteins/administration & dosage , Fungal Proteins/chemistry , Fungal Proteins/pharmacology , Leishmania/cytology , Leishmania/metabolism , Macrophages, Peritoneal/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C
12.
Mol Pharm ; 6(3): 813-24, 2009.
Article in English | MEDLINE | ID: mdl-19317431

ABSTRACT

Leishmaniasis is a human parasitic disease caused by infection by the protozoan Leishmania spp. Chemotherapy is currently the only treatment available, but its efficacy is increasingly challenged by the rising incidence of resistance and the frequent severe side effects associated with first-line drugs. Thus the development of leads with distinct mechanisms of action is urgently needed. A strategy often used for this purpose consists of assaying for leishmanicidal activity drugs formerly developed for other applications, such as amphotericin B (antifungal) or miltefosine (antitumor), among others, to profit from previous pharmacological and toxicological studies. Kahalalide F (KF) is a tumoricidal cyclic depsipeptide currently under phase II clinical trials for several types of cancer and psoriasis. Its mechanism of action has not been fully elucidated. Here we report the leishmanicidal activity of KF and its synthetic analogues at a micromolar range of concentrations. Its lethality is strongly linked to the alteration of the plasma membrane (PM) of the parasite based on (i) a rapid depolarization of the PM and uptake of the vital dye SYTOX Green upon its addition; (ii) evidence of severe morphological damage to the membrane of the parasite, as shown by transmission electron microscopy; and (iii) a rapid drop in the intracellular ATP levels, which correlates significantly with the leishmanicidal activity for active analogues, some of them with significant improvement of their therapeutic index with respect to the parental molecule. In addition to the basic knowledge obtained, this class of lethal mechanism is considerably less prone to the induction of resistance than classical drugs. All together, these observations foster further studies for the optimization of KF and its analogues as new anti-Leishmania leads with a new mode of action.


Subject(s)
Antineoplastic Agents/pharmacology , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Depsipeptides/pharmacology , Depsipeptides/therapeutic use , Leishmania/drug effects , Leishmaniasis/drug therapy , Animals , Antineoplastic Agents/chemistry , Antiprotozoal Agents/chemistry , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cells, Cultured , Clinical Trials as Topic , Depsipeptides/chemistry , Leishmania/ultrastructure , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Molecular Structure
13.
J Med Chem ; 50(24): 5994-6003, 2007 Nov 29.
Article in English | MEDLINE | ID: mdl-17973359

ABSTRACT

The leishmanicidal mechanism of miltefosine (hexadecylphosphocholine, MT) is not clearly understood. Valuable insights into its mode of action could be obtained by fluorescence techniques, given suitably emitting analogues. In this regard, the synthesis and biological characterization of two fully competent MT fluorescent analogues is reported here: all-(E)-13-phenyltrideca-6,8,10,12-tetraenylphosphocholine (PTE-MT) and all-(E)-13-phenyltrideca-8,10,12-trien-6-ynylphosphocholine (PTRI-MT). Both compounds show large absorption coefficients and a modest, but usable, fluorescence yield. Their activities were very similar to that of MT and were recognized by the MT uptake system of Leishmania. Their localization in living L. donovani promastigotes by confocal microscopy show a homogeneous intracellular distribution of the fluorescence. The concentration of PTRI-MT within the parasites (ca. 1.7 mM) showed a 100-fold enrichment relative to its external concentration. These results are consistent with a multiple target leishmanicidal mechanism for MT and validate the application of these analogues for pharmacokinetic and diagnostic studies concerning the chemotherapy of leishmaniasis.


Subject(s)
Fluorescent Dyes/chemical synthesis , Leishmania donovani/drug effects , Phosphorylcholine/analogs & derivatives , Trypanocidal Agents/chemical synthesis , Animals , Drug Resistance , Fluorescent Dyes/metabolism , Fluorescent Dyes/pharmacology , Leishmania donovani/metabolism , Microscopy, Confocal , Phosphorylcholine/chemical synthesis , Phosphorylcholine/metabolism , Phosphorylcholine/pharmacology , Stereoisomerism , Structure-Activity Relationship , Trypanocidal Agents/metabolism , Trypanocidal Agents/pharmacology
14.
Biochim Biophys Acta ; 1763(1): 110-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16377003

ABSTRACT

There is an increasing awareness of immune cell modulation by antimicrobial peptides. While this process often requires specific receptors for the peptides involved, several reports point out to a receptor-independent process. The cecropin A-melittin hybrid peptide CA(1-8)M(1-18) (KWKLFKKIGIGAVLKVLTTGLPALIS-amide) modifies gene expression in the macrophage line RAW 264.7 in the absence of any previous macrophage priming, suggesting a membrane permeation process. To further analyze the initial steps of this mechanism, we have studied the interaction of the peptide with these cells. Below 2 microM, CA(1-8)M(1-18) causes a concentration-dependent membrane depolarization partially reversible with time. At 2 microM, the accumulation of the SYTOX green vital dye is one half of that achieved with 0.05% Triton X-100. The binding level, as assessed by fluorescein-labeled CA(1-8)M(1-18), varies from 7.7+/-1.2 to 37.4+/-3.9 x 10(6) molecules/cell over a 0.5-4.0 microM concentration range. Electrophysiological experiments with 0.5 microM CA(1-8)M(1-18), a concentration that triggers maximal NOS2 expression and minimal toxicity, show a reversible current induction in the RAW 264.7 plasma membrane that is maintained as far as peptide is present. This activation of the macrophage involves the production of nitric oxide, a metabolite lethal for many pathogens that results from unspecific membrane permeation by antimicrobial peptides, and represents a new mode of action that may open new therapeutic possibilities for these compounds against intracellular pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cell Membrane Permeability/drug effects , Melitten/pharmacology , Monocytes/enzymology , Nitric Oxide Synthase Type II/biosynthesis , Peptides/pharmacology , Animals , Cells, Cultured , Electrophysiology , Enzyme Induction/drug effects , Fluorescent Dyes , Mice , Monocytes/cytology , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL