Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Mediators Inflamm ; 2023: 8387330, 2023.
Article in English | MEDLINE | ID: mdl-37101596

ABSTRACT

Theta- (θ-) defensins are pleiotropic host defense peptides with antimicrobial- and immune-modulating activities. Immune stimulation of cells with lipopolysaccharide (LPS, endotoxin) activates proinflammatory gene expression and cytokine secretion, both of which are attenuated by rhesus theta-defensin-1 (RTD-1) inhibition of NF-κB and MAP kinase pathways. Endotoxin tolerance is a condition that ensues when cells have an extended primary exposure to low levels of LPS, resulting in resistance to a subsequent LPS challenge. Recognition of LPS by Toll-like receptor-4 (TLR4) activates NF-κB, elevating levels of microRNA-146a (miR-146a), which targets IRAK1 and TRAF6 transcripts to reduce their protein levels and inhibits TLR signaling on secondary LPS stimulation. Here, we report that RTD-1 suppressed the expression of miR-146a and stabilized the IRAK1 protein in immune-stimulated, monocytic THP-1 cells. Cells that had primary exposure to LPS became endotoxin-tolerant, as evident from their failure to secrete TNF-α upon secondary endotoxin challenge. However, cells incubated with RTD-1 during the primary LPS stimulation secreted TNF-α after secondary LPS stimulation in an RTD-1 dose-dependent manner. Consistent with this, compared to the control treatment, cells treated with RTD-1 during primary LPS stimulation had increased NF-κB activity after secondary LPS stimulation. These results show that RTD-1 suppresses endotoxin tolerance by inhibiting the NF-κB pathway and demonstrates a novel inflammatory role for RTD-1 that is mediated by the downregulation of miR-146a during the innate immune response.


Subject(s)
MicroRNAs , NF-kappa B , NF-kappa B/metabolism , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha , Endotoxin Tolerance , Defensins , Endotoxins , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
2.
Physiol Genomics ; 51(12): 657-667, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31762409

ABSTRACT

Rhesus theta defensin-1 (RTD-1), a macrocyclic immunomodulatory host defense peptide from Old World monkeys, is therapeutic in pristane-induced arthritis (PIA) in rats, a model of rheumatoid arthritis (RA). RNA-sequence (RNA-Seq) analysis was used to interrogate the changes in gene expression in PIA rats, which identified 617 differentially expressed genes (DEGs) in PIA synovial tissue of diseased rats. Upstream regulator analysis showed upregulation of gene expression pathways regulated by TNF, IL1B, IL6, proinflammatory cytokines, and matrix metalloproteases (MMPs) involved in RA. In contrast, ligand-dependent nuclear receptors like the liver X-receptors NR1H2 and NR1H3 and peroxisome proliferator-activated receptor gamma (PPARG) were downregulated in arthritic synovia. Daily RTD-1 treatment of PIA rats for 1-5 days following disease presentation modulated 340 of the 617 disease genes, and synovial gene expression in PIA rats treated 5 days with RTD-1 closely resembled the gene signature of naive synovium. Systemic RTD-1 inhibited proinflammatory upstream regulators such as TNF, IL1, and IL6 and activated antiarthritic ligand-dependent nuclear receptor pathways, including PPARG, NR1H2, and NR1H3, that were suppressed in untreated PIA rats. RTD-1 also inhibited proinflammatory responses in IL-1ß-stimulated human RA fibroblast-like synoviocytes (FLS) in vitro and diminished expression of human orthologs of disease genes that are induced in rat PIA synovium. Thus, the antiarthritic mechanisms of systemic RTD-1 include homeostatic regulation of arthritogenic gene networks in a manner that correlates temporally with clinical resolution of rat PIA.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Fibroblasts/metabolism , Inflammation Mediators/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Synovial Membrane/metabolism , Transcriptome/drug effects , alpha-Defensins/pharmacology , alpha-Defensins/therapeutic use , Animals , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/metabolism , Cell Line , Cercopithecidae , Cytokines/genetics , Disease Models, Animal , Female , Humans , Immunosuppressive Agents/pharmacology , RNA-Seq , Rats , Synoviocytes/metabolism , Terpenes/pharmacology , Up-Regulation
3.
J Biol Chem ; 293(8): 2725-2734, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29317500

ABSTRACT

Theta-defensins (θ-defensins) are macrocyclic peptides expressed exclusively in granulocytes and selected epithelia of Old World monkeys. They contribute to anti-pathogen host defense responses by directly killing a diverse range of microbes. Of note, θ-defensins also modulate microbe-induced inflammation by affecting the production of soluble tumor necrosis factor (sTNF) and other proinflammatory cytokines. Here, we report that natural rhesus macaque θ-defensin (RTD) isoforms regulate sTNF cellular release by inhibiting TNF-α-converting enzyme (TACE; also known as adisintegrin and metalloprotease 17; ADAM17), the primary pro-TNF sheddase. Dose-dependent inhibition of cellular TACE activity by RTDs occurred when leukocytes were stimulated with live Escherichia coli cells as well as numerous Toll-like receptor agonists. Moreover, the relative inhibitory potencies of the RTD isoforms strongly correlated with their suppression of TNF release by stimulated blood leukocytes and THP-1 monocytes. RTD isoforms also inhibited ADAM10, a sheddase closely related to TACE. TACE inhibition was abrogated by introducing a single opening in the RTD-1 backbone, demonstrating that the intact macrocycle is required for enzyme inhibition. Enzymologic analyses showed that RTD-1 is a fast binding, reversible, non-competitive inhibitor of TACE. We conclude that θ-defensin-mediated inhibition of pro-TNF proteolysis by TACE represents a rapid mechanism for the regulation of sTNF and TNF-dependent inflammatory pathways. Molecules with structural and functional features mimicking those of θ-defensins may have clinical utility as TACE inhibitors for managing TNF-driven diseases.


Subject(s)
ADAM17 Protein/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Defensins/pharmacology , Leukocytes/drug effects , Monocytes/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Line , Chlorocebus aethiops , Colon/drug effects , Colon/immunology , Colon/metabolism , Defensins/chemistry , Escherichia coli/immunology , Escherichia coli/physiology , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Lipopolysaccharides/toxicity , Macaca mulatta , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Monocytes/immunology , Monocytes/metabolism , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/pharmacology , Proteolysis/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/metabolism
4.
Infect Immun ; 86(7)2018 07.
Article in English | MEDLINE | ID: mdl-29685982

ABSTRACT

Enteric α-defensins, termed cryptdins (Crps) in mice, and lysozymes secreted by Paneth cells contribute to innate host defense in the ileum. Antimicrobial factors, including lysozymes and ß-defensins, are often embedded in luminal glycosylated colonic Muc2 mucin secreted by goblet cells that form the protective mucus layer critical for gut homeostasis and pathogen invasion. In this study, we investigated ileal innate immunity against Entamoeba histolytica, the causative agent of intestinal amebiasis, by inoculating parasites in closed ileal loops in Muc2+/+ and Muc2-/- littermates and quantifying Paneth cell localization (lysozyme expression) and function (Crp secretion). Relative to Muc2+/+ littermates, Muc2-/- littermates showed a disorganized mislocalization of Paneth cells that was diffusely distributed, with elevated lysozyme secretion in the crypts and on villi in response to E. histolytica Inhibition of E. histolytica Gal/GalNAc lectin (Gal-lectin) binding with exogenous galactose and Entamoeba histolytica cysteine proteinase 5 (EhCP5)-negative E. histolytica had no effect on parasite-induced erratic Paneth cell lysozyme synthesis. Although the basal ileal expression of Crp genes was unaffected in Muc2-/- mice in response to E. histolytica, there was a robust release of proinflammatory cytokines and Crp peptide secretions in luminal exudates that was also present in the colon. Interestingly, E. histolytica-secreted cysteine proteinases cleaved the proregion of Crp4 but not the active form. These findings define Muc2 mucin as an essential component of ileal barrier function that regulates the localization and function of Paneth cells critical for host defense against microbes.


Subject(s)
Defensins/metabolism , Entamoeba histolytica/metabolism , Entamoeba histolytica/pathogenicity , Mucins/deficiency , Mucins/metabolism , Muramidase/metabolism , Paneth Cells/metabolism , Animals , Cell Proliferation/physiology , Host-Parasite Interactions , Humans , Mice
5.
Article in English | MEDLINE | ID: mdl-29610196

ABSTRACT

Systemic candidiasis is a growing health care concern that is becoming even more challenging due to the growing frequency of infections caused by multidrug-resistant (MDR) Candida species. Thus, there is an urgent need for new therapeutic approaches to candidiasis, including strategies bioinspired by insights into natural host defense against fungal pathogens. The antifungal properties of θ-defensins, macrocyclic peptides expressed in tissues of Old World monkeys, were investigated against a panel of drug-sensitive and drug-resistant clinical isolates of Candida albicans and non-albicans Candida species. Rhesus θ-defensin 1 (RTD-1), the prototype θ-defensin, was rapidly and potently fungicidal against drug-sensitive and MDR C. albicans strains. Fungal killing occurred by cell permeabilization that was temporally correlated with ATP release and intracellular accumulation of reactive oxygen species (ROS). Killing by RTD-1 was compared with that by histatin 5 (Hst 5), an extensively characterized anticandidal peptide expressed in human saliva. RTD-1 killed C. albicans much more rapidly and at a >200-fold lower concentration than that of Hst 5. Unlike Hst 5, the anticandidal activity of RTD-1 was independent of mitochondrial ATP production. Moreover, RTD-1 was completely resistant to Candida proteases for 2 h under conditions that rapidly and completely degraded Hst 5. MICs and minimum fungicidal concentrations (MFCs) of 14 natural θ-defensins isoforms against drug-resistant C. albicans isolates identified peptides that are more active than amphotericin B and/or caspofungin against fluconazole-resistant organisms, including MDR Candida auris. These results point to the potential of macrocyclic θ-defensins as structural templates for the design of antifungal therapeutics.


Subject(s)
Antifungal Agents/pharmacology , Candida/drug effects , Candidiasis/drug therapy , Defensins/pharmacology , Amphotericin B/pharmacology , Animals , Candida/isolation & purification , Candidiasis/microbiology , Caspofungin/pharmacology , Drug Resistance, Multiple, Fungal/physiology , Fluconazole/pharmacology , Histatins/pharmacology , Humans , Macaca mulatta , Microbial Sensitivity Tests , Protein Isoforms/pharmacology , Reactive Oxygen Species/metabolism
6.
PLoS Pathog ; 12(3): e1005474, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26933888

ABSTRACT

α-defensins are abundant antimicrobial peptides with broad, potent antibacterial, antifungal, and antiviral activities in vitro. Although their contribution to host defense against bacteria in vivo has been demonstrated, comparable studies of their antiviral activity in vivo are lacking. Using a mouse model deficient in activated α-defensins in the small intestine, we show that Paneth cell α-defensins protect mice from oral infection by a pathogenic virus, mouse adenovirus 1 (MAdV-1). Survival differences between mouse genotypes are lost upon parenteral MAdV-1 infection, strongly implicating a role for intestinal defenses in attenuating pathogenesis. Although differences in α-defensin expression impact the composition of the ileal commensal bacterial population, depletion studies using broad-spectrum antibiotics revealed no effect of the microbiota on α-defensin-dependent viral pathogenesis. Moreover, despite the sensitivity of MAdV-1 infection to α-defensin neutralization in cell culture, we observed no barrier effect due to Paneth cell α-defensin activation on the kinetics and magnitude of MAdV-1 dissemination to the brain. Rather, a protective neutralizing antibody response was delayed in the absence of α-defensins. This effect was specific to oral viral infection, because antibody responses to parenteral or mucosal ovalbumin exposure were not affected by α-defensin deficiency. Thus, α-defensins play an important role as adjuvants in antiviral immunity in vivo that is distinct from their direct antiviral activity observed in cell culture.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , Anti-Infective Agents/immunology , Antibodies, Neutralizing/immunology , Antiviral Agents/immunology , Defensins/immunology , Animals , Female , Humans , Ileum/immunology , Intestine, Small/immunology , Intestines/immunology , Male , Mice , Mice, Inbred C57BL , Paneth Cells/immunology , alpha-Defensins/immunology
7.
Biochemistry ; 55(27): 3784-93, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27305837

ABSTRACT

Chemokines make up a superfamily of ∼50 small secreted proteins (8-12 kDa) involved in a host of physiological processes and disease states, with several previously shown to have direct antimicrobial activity comparable to that of defensins in efficacy. XCL1 is a unique metamorphic protein that interconverts between the canonical chemokine fold and a novel all-ß-sheet dimer. Phylogenetic analysis suggests that, within the chemokine family, XCL1 is most closely related to CCL20, which exhibits antibacterial activity. The in vitro antimicrobial activity of WT-XCL1 and structural variants was quantified using a radial diffusion assay (RDA) and in solution bactericidal assays against Gram-positive and Gram-negative species of bacteria. Comparisons of WT-XCL1 with variants that limit metamorphic interconversion showed a loss of antimicrobial activity when restricted to the conserved chemokine fold. These results suggest that metamorphic folding of XCL1 is required for potent antimicrobial activity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Chemokines, C/pharmacology , Protein Folding , Amino Acid Sequence , Humans , Phylogeny , Protein Binding , Sequence Homology, Amino Acid
8.
Infect Immun ; 82(6): 2195-202, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24614658

ABSTRACT

Mammalian α-defensins are approximately 4- to 5-kDa broad-spectrum antimicrobial peptides and abundant granule constituents of neutrophils and small intestinal Paneth cells. The bactericidal activities of amphipathic α-defensins depend in part on electropositive charge and on hydrophobic amino acids that enable membrane disruption by interactions with phospholipid acyl chains. Alignment of α-defensin primary structures identified conserved hydrophobic residues in the loop formed by the Cys(III)-Cys(V) disulfide bond, and we have studied their role by testing the effects of mutagenesis on bactericidal activities. Mouse α-defensin 4 (Crp-4) and rhesus myeloid α-defensin 4 (RMAD-4) were selected for these studies, because they are highly bactericidal in vitro and have the same overall electropositive charge. Elimination of hydrophobicity by site-directed mutagenesis at those positions in Crp-4 attenuated bactericidal activity markedly. In contrast to native Crp-4, the (I23/F25/L26/G)-Crp-4 variant lacked bactericidal activity against Salmonella enterica serovar Typhimurium and did not permeabilize Escherichia coli ML35 cells as a result of removing aliphatic side chains by Gly substitutions. Ala replacements in (I23/F25/L26/A)-Crp-4 restored activity, evidence that hydrophobicity contributed by Ala methyl R-groups was sufficient for activity. In macaques, neutrophil α-defensin RMAD-6 is identical to RMAD-4, except for a F28S difference, and (F28S)-RMAD-4 mutagenesis attenuated RMAD-4 bactericidal activity and E. coli permeabilization. Interestingly, (R31/32D)-Crp-4 lacks activity in these assays despite the presence of the Ile23, Phe25, and Leu26 hydrophobic patch. We infer that electrostatic interactions between cationic α-defensin residues and negative charge on bacteria precede interactions between critical hydrophobic residue positions that mediate membrane disruption and bacterial cell killing.


Subject(s)
Bacteria/drug effects , alpha-Defensins/pharmacology , Amino Acid Substitution , Animals , Cell Membrane Permeability/drug effects , Cells, Cultured , Hydrophobic and Hydrophilic Interactions , Macaca mulatta , Mice , Mutagenesis, Site-Directed , Recombinant Proteins/pharmacology , alpha-Defensins/chemistry
9.
J Immunol ; 189(11): 5421-33, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23109723

ABSTRACT

Acute kidney injury (AKI) is frequently complicated by extrarenal multiorgan injury, including intestinal and hepatic dysfunction. In this study, we hypothesized that a discrete intestinal source of proinflammatory mediators drives multiorgan injury in response to AKI. After induction of AKI in mice by renal ischemia-reperfusion or bilateral nephrectomy, small intestinal Paneth cells increased the synthesis and release of IL-17A in conjunction with severe intestinal apoptosis and inflammation. We also detected significantly increased IL-17A in portal and systemic circulation after AKI. Intestinal macrophages appear to transport released Paneth cell granule constituents induced by AKI, away from the base of the crypts into the liver. Genetic or pharmacologic depletion of Paneth cells decreased small intestinal IL-17A secretion and plasma IL-17A levels significantly and attenuated intestinal, hepatic, and renal injury after AKI. Similarly, portal delivery of IL-17A in macrophage-depleted mice decreased markedly. In addition, intestinal, hepatic, and renal injury following AKI was attenuated without affecting intestinal IL-17A generation. In conclusion, AKI induces IL-17A synthesis and secretion by Paneth cells to initiate intestinal and hepatic injury by hepatic and systemic delivery of IL-17A by macrophages. Modulation of Paneth cell dysregulation may have therapeutic implications by reducing systemic complications arising from AKI.


Subject(s)
Acute Kidney Injury/pathology , Inflammation/pathology , Kidney/pathology , Lung/pathology , Paneth Cells/pathology , Reperfusion Injury/pathology , Acute Kidney Injury/complications , Acute Kidney Injury/immunology , Animals , Apoptosis , Inflammation/complications , Inflammation/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Kidney/immunology , Lung/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Nephrectomy , Paneth Cells/immunology , Portal System/immunology , Reperfusion Injury/complications , Reperfusion Injury/immunology
10.
J Immunol ; 188(12): 6399-406, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22611239

ABSTRACT

The mucosal immune network is a crucial barrier preventing pathogens from entering the body. The network of immune cells that mediates the defensive mechanisms in the mucosa is likely shaped by chemokines, which attract a wide range of immune cells to specific sites of the body. Chemokines have been divided into homeostatic or inflammatory depending upon their expression patterns. Additionally, several chemokines mediate direct killing of invading pathogens, as exemplified by CCL28, a mucosa-associated chemokine that exhibits antimicrobial activity against a range of pathogens. CXCL17 was the last chemokine ligand to be described and is the 17th member of the CXC chemokine family. Its expression pattern in 105 human tissues and cells indicates that CXCL17 is a homeostatic, mucosa-associated chemokine. Its strategic expression in mucosal tissues suggests that it is involved in innate immunity and/or sterility of the mucosa. To test the latter hypothesis, we tested CXCL17 for possible antibacterial activity against a panel of pathogenic and opportunistic bacteria. Our results indicate that CXCL17 has potent antimicrobial activities and that its mechanism of antimicrobial action involves peptide-mediated bacterial membrane disruption. Because CXCL17 is strongly expressed in bronchi, we measured it in bronchoalveolar lavage fluids and observed that it is strongly upregulated in idiopathic pulmonary fibrosis. We conclude that CXCL17 is an antimicrobial mucosal chemokine that may play a role in the pathogenesis of interstitial lung diseases.


Subject(s)
Anti-Bacterial Agents/immunology , Chemokines, CXC/immunology , Idiopathic Pulmonary Fibrosis/immunology , Immunity, Innate/immunology , Respiratory Mucosa/immunology , Aged , Anti-Bacterial Agents/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Chemokines, CXC/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Immunohistochemistry , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/chemistry , Respiratory Mucosa/metabolism
11.
J Biol Chem ; 287(14): 11205-12, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22334698

ABSTRACT

Paneth cell α-defensins mediate host defense and homeostasis at the intestinal mucosal surface. In mice, matrix metalloproteinase-7 (MMP7) converts inactive pro-α-defensins (proCrps) to bactericidal forms by proteolysis at specific proregion cleavage sites. MMP7(-/-) mice lack mature α-defensins in Paneth cells, accumulating unprocessed precursors for secretion. To test for activation of secreted pro-α-defensins by host and microbial proteinases in the absence of MMP7, we characterized colonic luminal α-defensins. Protein extracts of complete (organ plus luminal contents) ileum, cecum, and colon of MMP7-null and wild-type mice were analyzed by sequential gel permeation chromatography/acid-urea polyacrylamide gel analyses. Mature α-defensins were identified by N-terminal sequencing and mass spectrometry and characterized in bactericidal assays. Abundance of specific bacterial groups was measured by qPCR using group specific 16 S rDNA primers. Intact, native α-defensins, N-terminally truncated α-defensins, and α-defensin variants with novel N termini due to alternative processing were identified in MMP7(-/-) cecum and colon, and proteinases of host and microbial origin catalyzed proCrp4 activation in vitro. Although Paneth cell α-defensin deficiency is associated with ileal microbiota alterations, the cecal and colonic microbiota of MMP7(-/-) and wild-type mice were not significantly different. Thus, despite the absence of MMP7, mature α-defensins are abundant in MMP7(-/-) cecum and colon due to luminal proteolytic activation by alternative host and microbial proteinases. MMP7(-/-) mice only lack processed α-defensins in the small intestine, and the model is not appropriate for studying effects of α-defensin deficiency in cecal or colonic infection or disease.


Subject(s)
Paneth Cells/metabolism , alpha-Defensins/metabolism , Amino Acid Sequence , Animals , Bacteria/enzymology , Cecum/cytology , Cecum/microbiology , Colon/cytology , Colon/microbiology , Matrix Metalloproteinase 7/deficiency , Metagenome , Mice , Molecular Sequence Data , Paneth Cells/microbiology , Peptide Hydrolases/metabolism , alpha-Defensins/chemistry
12.
J Biol Chem ; 287(26): 21866-72, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22566697

ABSTRACT

The conserved tridisulfide array of the α-defensin family imposes a common triple-stranded ß-sheet topology on peptides that may have highly diverse primary structures, resulting in differential outcomes after targeted mutagenesis. In mouse cryptdin-4 (Crp4) and rhesus myeloid α-defensin-4 (RMAD4), complete substitutions of Arg with Lys affect bactericidal peptide activity very differently. Lys-for-Arg mutagenesis attenuates Crp4, but RMAD4 activity remains mostly unchanged. Here, we show that the differential biological effect of Lys-for-Arg replacements can be understood by the distinct phase behavior of the experimental peptide-lipid system. In Crp4, small-angle x-ray scattering analyses showed that Arg-to-Lys replacements shifted the induced nanoporous phases to a different range of lipid compositions compared with the Arg-rich native peptide, consistent with the attenuation of bactericidal activity by Lys-for-Arg mutations. In contrast, such phases generated by RMAD4 were largely unchanged. The concordance between small-angle x-ray scattering measurements and biological activity provides evidence that specific types of α-defensin-induced membrane curvature-generating tendencies correspond directly to bactericidal activity via membrane destabilization.


Subject(s)
Arginine/metabolism , Protein Precursors/metabolism , alpha-Defensins/metabolism , Animals , Anti-Infective Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Arginine/chemistry , Defensins/chemistry , Escherichia coli/metabolism , Immunity, Innate , Lipids/chemistry , Lysine/chemistry , Mice , Normal Distribution , Peptides/chemistry , Scattering, Radiation , X-Rays , alpha-Defensins/chemistry
14.
Antimicrob Agents Chemother ; 57(4): 1823-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23380721

ABSTRACT

Cationic antimicrobial peptides are essential components of the innate immune system. As a major family of mammalian antimicrobial peptides, defensins are expressed mainly by mucosal epithelial cells and promyelocytes. Despite the capacity to kill a broad spectrum of bacteria through physical disruption of membranes, most defensins show substantially reduced antibacterial activities in the presence of monovalent and divalent cations, thereby limiting their therapeutic potential, particularly for the treatment of systemic infections. Genome-wide computational screening of the rat genome led to the identification of the gene for a novel α-defensin-related peptide that we termed rattusin. Rattusin shares a highly conserved signal and prosequence with mammalian α-defensins, but instead of the canonical α-defensin six-cysteine motif, rattusin consists of five cysteines with a distinctive spacing pattern. Furthermore, rattusin is preferentially expressed in Paneth cells of the distal small intestine with potent antibacterial activity against a broad range of Gram-negative and Gram-positive bacteria, including antibiotic-resistant strains. The MICs were mostly in the range of 2 to 4 µM, with no appreciable toxicity to mammalian cells at up to 100 µM. In contrast to classical α- and ß-defensins, rattusin retained its activity in the presence of physiological concentrations of NaCl and Mg(2+), making it an attractive antimicrobial candidate for both topical and systemic applications.


Subject(s)
Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Peptides/metabolism , Peptides/pharmacology , alpha-Defensins/chemistry , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Caco-2 Cells , Cysteine/chemistry , Cysteine/genetics , Humans , In Situ Hybridization , Intestine, Small/metabolism , Klebsiella pneumoniae/drug effects , Listeria monocytogenes/drug effects , Molecular Sequence Data , Peptides/chemistry , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Staphylococcus aureus/drug effects , beta-Defensins/chemistry
15.
J Immunol ; 186(3): 1589-97, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21178012

ABSTRACT

Gastrointestinal mucosa is an early target of HIV and a site of viral replication and severe CD4(+) T cell depletion. However, effects of HIV infection on gut mucosal innate immune defense have not been fully investigated. Intestinal Paneth cell-derived α-defensins constitute an integral part of the gut mucosal innate defense against microbial pathogens. Using the SIV-infected rhesus macaque model of AIDS, we examined the level of expression of rhesus enteric α-defensins (REDs) in the jejunal mucosa of rhesus macaques during all stages of SIV infection using real-time PCR, in situ hybridization, and immunohistochemistry. An increased expression of RED mRNAs was found in PC at the base of the crypts in jejunum at all stages of SIV infection as compared with uninfected controls. This increase correlated with active viral replication in gut-associated lymphoid tissue. Loss of RED protein accumulation in PC was seen in animals with simian AIDS. This was associated with the loss of secretory granules in PC, suggesting an increase in degranulation during advanced SIV disease. The α-defensin-mediated innate mucosal immunity was maintained in PC throughout the course of SIV infection despite the mucosal CD4(+) T cell depletion. The loss of RED protein accumulation and secretion was associated with an increased incidence of opportunistic enteric infections and disease progression. Our findings suggest that local innate immune defense exerted by PC-derived defensins contributes to the protection of gut mucosa from opportunistic infections during the course of SIV infection.


Subject(s)
Gene Expression Regulation, Viral/immunology , Immunity, Innate , Intestinal Mucosa/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , alpha-Defensins/biosynthesis , Animals , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Count , Disease Models, Animal , Gene Expression Regulation, Viral/drug effects , Immunity, Innate/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Jejunum/drug effects , Jejunum/immunology , Jejunum/pathology , Longitudinal Studies , Lymphocyte Depletion , Macaca mulatta , Paneth Cells/drug effects , Paneth Cells/immunology , Paneth Cells/pathology , RNA, Messenger/biosynthesis , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus/drug effects , Up-Regulation/drug effects , Up-Regulation/immunology , alpha-Defensins/genetics , alpha-Defensins/physiology
16.
Sci Rep ; 13(1): 3953, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36894646

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a chronic liver disease characterized by fibrosis that develops from fatty liver. Disruption of intestinal microbiota homeostasis, dysbiosis, is associated with fibrosis development in NASH. An antimicrobial peptide α-defensin secreted by Paneth cells in the small intestine is known to regulate composition of the intestinal microbiota. However, involvement of α-defensin in NASH remains unknown. Here, we show that in diet-induced NASH model mice, decrease of fecal α-defensin along with dysbiosis occurs before NASH onset. When α-defensin levels in the intestinal lumen are restored by intravenous administration of R-Spondin1 to induce Paneth cell regeneration or by oral administration of α-defensins, liver fibrosis is ameliorated with dissolving dysbiosis. Furthermore, R-Spondin1 and α-defensin improved liver pathologies together with different features in the intestinal microbiota. These results indicate that decreased α-defensin secretion induces liver fibrosis through dysbiosis, further suggesting Paneth cell α-defensin as a potential therapeutic target for NASH.


Subject(s)
Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease , alpha-Defensins , Animals , Mice , Amino Acids , Choline , Diet, High-Fat/methods , Dysbiosis/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Paneth Cells/pathology
17.
Cell Mol Gastroenterol Hepatol ; 16(1): 39-62, 2023.
Article in English | MEDLINE | ID: mdl-37030630

ABSTRACT

BACKGROUND & AIMS: Loss-of-function variants in the PTPN2 gene are associated with increased risk of inflammatory bowel disease. We recently showed that Ptpn2 is critical for intestinal epithelial cell (IEC) barrier maintenance, IEC-macrophage communication, and modulation of the gut microbiome in mice, restricting expansion of a small intestinal pathobiont associated with inflammatory bowel disease. Here, we aimed to identify how Ptpn2 loss affects ileal IEC subtypes and their function in vivo. METHODS: Constitutive Ptpn2 wild-type, heterozygous, and knockout (KO) mice, as well as mice with inducible deletion of Ptpn2 in IECs, were used in the study. Investigation was performed using imaging techniques, flow cytometry, enteroid culture, and analysis of gene and protein levels of IEC markers. RESULTS: Partial transcriptome analysis showed that expression of Paneth cell-associated antimicrobial peptides Lyz1, Pla2g2a, and Defa6 was down-regulated markedly in Ptpn2-KO mice compared with wild-type and heterozygous. In parallel, Paneth cell numbers were reduced, their endoplasmic reticulum architecture was disrupted, and the endoplasmic reticulum stress protein, C/EBP-homologous protein (CHOP), was increased in Ptpn2-KO mice. Despite reduced Paneth cell number, flow cytometry showed increased expression of the Paneth cell-stimulatory cytokines interleukin 22 and interferon γ+ in CD4+ T cells isolated from Ptpn2-KO ileum. Key findings in constitutive Ptpn2-KO mice were confirmed in epithelium-specific Ptpn2ΔIEC mice, which also showed impaired lysozyme protein levels in Paneth cells compared with Ptpn2fl/fl control mice. CONCLUSIONS: Constitutive Ptpn2 deficiency affects Paneth cell viability and compromises Paneth cell-specific antimicrobial peptide production. The observed effects may contribute to the increased susceptibility to intestinal infection and dysbiosis in these mice.


Subject(s)
Inflammatory Bowel Diseases , Paneth Cells , Mice , Animals , Paneth Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Cell Survival , Inflammatory Bowel Diseases/genetics , Ileum/metabolism , Mice, Knockout
18.
Amino Acids ; 43(4): 1471-83, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22286872

ABSTRACT

Salt-bridge interactions between acidic and basic amino acids contribute to the structural stability of proteins and to protein-protein interactions. A conserved salt-bridge is a canonical feature of the α-defensin antimicrobial peptide family, but the role of this common structural element has not been fully elucidated. We have investigated mouse Paneth cell α-defensincryptdin-4 (Crp4) and peptide variants with mutations at Arg7 or Glu15 residue positions to disrupt the salt-bridge and assess the consequences on Crp4 structure, function, and stability. NMR analyses showed that both (R7G)-Crp4 and (E15G)-Crp4 adopt native-like structures, evidence of fold plasticity that allows peptides to reshuffle side chains and stabilize the structure in the absence of the salt-bridge. In contrast, introduction of a large hydrophobic side chain at position 15, as in (E15L)-Crp4 cannot be accommodated in the context of the Crp4 primary structure. Regardless of which side of the salt-bridge was mutated, salt-bridge variants retained bactericidal peptide activity with differential microbicidal effects against certain bacterial cell targets, confirming that the salt-bridge does not determine bactericidal activity per se. The increased structural flexibility induced by salt-bridge disruption enhanced peptide sensitivity to proteolysis. Although sensitivity to proteolysis by MMP7 was unaffected by most Arg(7) and Glu(150 substitutions, every salt-bridge variant was degraded extensively by trypsin. Moreover, the salt-bridge facilitates adoption of the characteristic α-defensin fold as shown by the impaired in vitro refolding of (E15D)-proCrp4, the most conservative salt-bridge disrupting replacement. In Crp4, therefore, the canonical α-defensin salt-bridge facilitates adoption of the characteristic α-defensin fold, which decreases structural flexibility and confers resistance todegradation by proteinases.


Subject(s)
Anti-Infective Agents/chemistry , alpha-Defensins/chemistry , Amino Acid Sequence , Animals , Anti-Infective Agents/pharmacology , Arginine/chemistry , Arginine/genetics , Glutamic Acid/chemistry , Glutamic Acid/genetics , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/growth & development , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Matrix Metalloproteinase 7/chemistry , Mice , Microbial Viability/drug effects , Molecular Sequence Data , Mutation , Paneth Cells/physiology , Protein Stability , Protein Structure, Secondary , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Salts , Trypsin/chemistry , alpha-Defensins/genetics , alpha-Defensins/pharmacology
19.
Cell Mol Life Sci ; 68(13): 2215-29, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21560070

ABSTRACT

Paneth cells at the base of small intestinal crypts of Lieberkühn secrete high levels of α-defensins in response to cholinergic and microbial stimuli. Paneth cell α-defensins are broad spectrum microbicides that function in the extracellular environment of the intestinal lumen, and they are responsible for the majority of secreted bactericidal peptide activity. Paneth cell α-defensins confer immunity to oral infection by Salmonella enterica serovar Typhimurium, and they are major determinants of the composition of the small intestinal microbiome. In addition to host defense molecules such as α-defensins, lysozyme, and Pla2g2a, Paneth cells also produce and release proinflammatory mediators as components of secretory granules. Disruption of Paneth cell homeostasis, with subsequent induction of endoplasmic reticulum stress, autophagy, or apoptosis, contributes to inflammation in diverse genetic and experimental mouse models.


Subject(s)
Paneth Cells/immunology , alpha-Defensins/metabolism , Amino Acid Sequence , Animals , Gene Expression , Host-Pathogen Interactions , Metagenome/immunology , Mice , Molecular Sequence Data , Paneth Cells/microbiology , Protein Conformation , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Secretory Vesicles/immunology , alpha-Defensins/chemistry , alpha-Defensins/genetics
20.
Biochemistry ; 50(48): 10508-19, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22040603

ABSTRACT

Defensins are antimicrobial peptides that are important in the innate immune defense of mammals. Upon stimulation by bacterial antigens, enteric α-defensins are secreted into the intestinal lumen where they have potent microbicidal activities. Cryptdin-4 (Crp4) is an α-defensin expressed in Paneth cells of the mouse small intestine and the most bactericidal of the known cryptdin isoforms. The structure of Crp4 consists of a triple-stranded antiparallel ß-sheet but lacks three amino acids between the fourth and fifth cysteine residues, making them distinct from other α-defensins. The structure also reveals that the α-amino and C-terminal carboxylic groups are in the proximity of each other (d ≈ 3 Å) in the folded structure. We present here the biosynthesis of backbone-cyclized Crp4 using a modified protein splicing unit or intein. Our data show that cyclized Crp4 can be biosynthesized by using this approach both in vitro and in vivo, although the expression yield was significantly lower when the protein was produced inside the cell. The resulting cyclic defensins retained the native α-defensin fold and showed equivalent or better microbicidal activities against several Gram-positive and Gram-negative bacteria when compared to native Crp4. No detectable hemolytic activity against human red blood cells was observed for either native Crp4 or its cyclized variants. Moreover, both forms of Crp4 also showed high stability to degradation when incubated with human serum. Altogether, these results indicate the potential for backbone-cyclized defensins in the development of novel peptide-based antimicrobial compounds.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/pharmacology , Peptides, Cyclic/biosynthesis , Peptides, Cyclic/pharmacology , Protein Conformation , alpha-Defensins/biosynthesis , alpha-Defensins/physiology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/blood , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , Hemolysis/drug effects , Humans , Mice , Molecular Sequence Data , Paneth Cells/chemistry , Paneth Cells/metabolism , Paneth Cells/microbiology , Peptides, Cyclic/blood , Protein Folding , Protein Stability , alpha-Defensins/blood
SELECTION OF CITATIONS
SEARCH DETAIL