Article

Defensins enable macrophages to inhibit the intracellular proliferation of Listeria monocytogenes

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

Listeria monocytogenes is a facultative intracellular pathogen that infects a large diversity of host cells, including macrophages. To avoid the phagosome microbicidal environment, L. monocytogenes secretes a pore-forming toxin (listeriolysin O, LLO) that releases the bacterium into the cytoplasm. We hypothesized that the α-defensins (HNPs) and/or humanized θ-defensin (RC-1) peptides produced by human and non-human primate neutrophils, respectively, cooperate with macrophages to control L. monocytogenes infection. Our results establish that HNP-1 and RC-1 enable macrophages to control L. monocytogenes intracellular growth by inhibiting phagosomal escape, as a consequence, bacteria remain trapped in a LAMP-1-positive phagosome. Importantly, HNP-1 interaction with macrophages and RC-1 interaction with bacteria are required to prevent macrophage infection. In accordance with these results, RC-1 is a more potent anti-listerial peptide than HNP-1 and HNP-1 is acquired by macrophages and trafficked to the phagocytosed bacteria. Finally, HNP-1 and RC-1 antimicrobial activity is complemented by their ability to prevent LLO function through two mechanisms, blocking LLO-dependent perforation of macrophage membranes and the release of LLO from the bacteria. In conclusion, at the site of infection the cooperation between antimicrobial peptides, such as HNP-1, and macrophages likely plays a critical role in the innate immune defence against L. monocytogenes.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... Compared to their parent peptide (plectasin), both analogs effectively reduced the intracellular bacterial load . Other defensinlike AMPs, including HNP-1 (a-human defensins) and RC-1 (humanized q-defensin, retrocyclin -1), were tested against intracellular Listeria monocytogenes (Arnett et al., 2011). All these peptides caused a dose-dependent inhibition of bacterial proliferation, with more promising results for RC-1 ( Table 1) (Arnett et al., 2011). ...
... Other defensinlike AMPs, including HNP-1 (a-human defensins) and RC-1 (humanized q-defensin, retrocyclin -1), were tested against intracellular Listeria monocytogenes (Arnett et al., 2011). All these peptides caused a dose-dependent inhibition of bacterial proliferation, with more promising results for RC-1 ( Table 1) (Arnett et al., 2011). ...
... In vitro (Arnett et al., 2011;Witter et al., 2016;Lacoma et al., 2017) a-defensin HNP-1 Listeria monocytogenes ...
Article
Full-text available
Bacterial infections caused by intracellular pathogens are difficult to control. Conventional antibiotic therapies are often ineffective, as high doses are needed to increase the number of antibiotics that will cross the host cell membrane to act on the intracellular bacterium. Moreover, higher doses of antibiotics may lead to elevated severe toxic effects against host cells. In this context, antimicrobial peptides (AMPs) and cell-penetrating peptides (CPPs) have shown great potential to treat such infections by acting directly on the intracellular pathogenic bacterium or performing the delivery of cargos with antibacterial activities. Therefore, in this mini-review, we cover the main AMPs and CPPs described to date, aiming at intracellular bacterial infection treatment. Moreover, we discuss some of the proposed mechanisms of action for these peptide classes and their conjugation with other antimicrobials.
... Indeed, LLO-deficient L. monocytogenes strains remain trapped in the endocytic vesicle, are unable to divide intracellularly, and are nonvirulent in vivo (6,10). LLO mediates intracellular survival of L. monocytogenes in primary cells and immortalized cell lines from various animal species (1,(11)(12)(13). However, activated macrophages display increased resistance to infection by L. monocytogenes (14). ...
... Twenty-four hours before the experiment, 10 5 cells/well were plated in 24-well tissue culturetreated dishes coated with 30 mg fibronectin. Wild-type (wt; DP10403S), isogenic LLO-deficient (Dhly, DP-L2161), and LLO-overexpressing [LLO + ; DP10403S harboring the pAM401 plasmid encoding LLO that secretes 5.5 6 1.3-fold more LLO than wt (12)] L. monocytogenes and L. innocua (ATCC 33090; American Type Culture Collection) were cultured overnight in brain heart infusion (BHI) at 37˚C with shaking. The following day, bacteria were diluted 1/20 in BHI and grown at 37˚C until OD at 600 nm reached 0.7-0.8. ...
... In addition to their phagocytic role, neutrophils may cooperate with other cell types to limit their infection. For example, neutrophils produce inflammatory mediators (65) and release antimicrobial molecules that can be taken up by other cells to reinforce their defense mechanisms (12,(77)(78)(79). LLO promotes L. monocytogenes intracellular survival in several cell types, but appears to exacerbate the antimicrobial responses of neutrophils. ...
Article
Full-text available
The pore-forming toxin listeriolysin O (LLO) is a major virulence factor secreted by the facultative intracellular pathogen Listeria monocytogenes. This toxin facilitates L. monocytogenes intracellular survival in macrophages and diverse nonphagocytic cells by disrupting the internalization vesicle, releasing the bacterium into its replicative niche, the cytosol. Neutrophils are innate immune cells that play an important role in the control of infections, yet it was unknown if LLO could confer a survival advantage to L. monocytogenes in neutrophils. We report that LLO can enhance the phagocytic efficiency of human neutrophils and is unable to protect L. monocytogenes from intracellular killing. To explain the absence of L. monocytogenes survival in neutrophils, we hypothesized that neutrophil degranulation leads to the release of LLO-neutralizing molecules in the forming phagosome. In support of this, L. monocytogenes is a potent inducer of neutrophil degranulation, since its virulence factors, such as LLO, facilitate granule exocytosis. Within the first few minutes of interaction with L. monocytogenes, granules can fuse with the plasma membrane at the bacterial interaction site before closure of the phagosome. Furthermore, granule products directly degrade LLO, irreversibly inhibiting its activity. The matrix metalloproteinase-8, stored in secondary granules, was identified as an endoprotease that degrades LLO, and blocking neutrophil proteases increased L. monocytogenes intracellular survival. In conclusion, we propose that LLO degradation by matrix metalloproteinase-8 during phagocytosis protects neutrophil membranes from perforation and contributes to maintaining L. monocytogenes in a bactericidal phagosome from which it cannot escape.
... LAB have been demonstrated to increase Paneth cell number and/or stimulate AMPs, depending on the type of LAB strains and epithelial environment context [89,96]. AMPs such as α-defensin and cathelin-related antimicrobial peptides have been demonstrated to provide effective protection to host cells against L. monocytogenes and exert a synergistic effect with macrophages to limit the secretion of LLO and subsequent intracellular proliferation of L. monocytogenes [113,114]. Cathelicidins and defensin bind to the phospholipids group of Listeria membrane, e.g., lipid II through electrostatic force can create pores, e.g., Human Neutrophil α-defensin 2 dimer forms multimetric pores, impedes peptidoglycan formation, and alters Listeria integrity [41,49,112,115,116]. The release of AMPs is partially dependent on NOD2 [49,110], the molecule particularly highly expressed on Paneth cells, and thus was suggested to be crucial to regulate Paneth cells' antimicrobial activity [96,117], or initiated by TLRs [115]. ...
... However, the exact role of NOD in AMPs' expression is poorly known because there are conflicting reports, suggesting the possibility of an additional function of NOD in Paneth cells beyond regulation of AMPs' secretion and activity [43,48,90,115,117,120]. AMPs perform different functions through different mechanisms in exerting an antipathogenic effect, including enzymatic or nonenzymatic reaction, or interaction with the intracellular component to inhibit DNA and protein synthesis, and protein folding [113]. In short, every AMP plays a crucial and specific role in protecting host cells from pathogenic infection [43,[114][115][116]. ...
Article
Full-text available
Listeriosis is a severe food borne disease with a mortality rate of up to 30% caused by pathogenic Listeria monocytogenes via the production of several virulence factors including listeriolysin O (LLO), transcriptional activator (PrfA), actin (Act), internalin (Int), etc. It is a foodborne disease predominantly causing infections through consumption of contaminated food and is often associated with ready-to-eat food (RTE) and dairy products. Common medication for listeriosis such as antibiotics might cause an eagle effect and antibiotic resistance if it is overused. Therefore, exploration of the use of lactic acid bacteria (LAB) with probiotic characteristics and multiple antimicrobial properties is increasingly getting attention for their capability to treat listeriosis, vaccine development, and hurdle technologies. The antilisterial gene, a gene coding to produce antimicrobial peptide (AMP), one of the inhibitory substances found in LAB, is one of the potential key factors in listeriosis treatment, coupled with the vast array of functions and strategies; this review summarizes the various strategies by LAB against L. monocytogenes and the prospect in development of a ‘generally regarded as safe’ LAB for treatment of listeriosis.
... A different mechanism underlies the effect of the alpha defensin HNP-1, which was found to decrease Listeria monocytogenes proliferation inside murine bone-marrow-derived macrophages by preventing the release and activity of listeriolysin O (LLO) [83]. In order to proliferate, Listeria relies on LLO, amongst other mechanisms, to disrupt the phagosomal membrane and escape to the macrophage cytosol where it will proliferate and invade neighbor cells. ...
... In order to proliferate, Listeria relies on LLO, amongst other mechanisms, to disrupt the phagosomal membrane and escape to the macrophage cytosol where it will proliferate and invade neighbor cells. By impairing the activity of LLO, HNP-1 was able to prevent bacterial escape from the phagosomal harmful environment, cooperating with the macrophage antimicrobial mechanisms to control bacterial proliferation and dissemination [83]. ...
Article
Full-text available
The treatment of infectious diseases is increasingly prone to failure due to the rapid spread of antibiotic-resistant pathogens. Antimicrobial peptides (AMPs) are natural components of the innate immune system of most living organisms. Their capacity to kill microbes through multiple mechanisms makes the development of bacterial resistance less likely. Additionally, AMPs have important immunomodulatory effects, which critically contribute to their role in host defense. In this paper, we review the most recent evidence for the importance of AMPs in host defense against intracellular pathogens, particularly intra-macrophagic pathogens, such as mycobacteria. Cathelicidins and defensins are reviewed in more detail, due to the abundance of studies on these molecules. The cell-intrinsic as well as the systemic immune-related effects of the different AMPs are discussed. In the face of the strong potential emerging from the reviewed studies, the prospects for future use of AMPs as part of the therapeutic armamentarium against infectious diseases are presented.
... Due to the central role LLO plays during pathogenesis, molecular strategies preventing its synthesis, secretion, and/or activity are of great medical interest. For example, human antimicrobial peptides that belong to the α-defensin family, such as HNP1, prevent both LLO release by the bacterium and LLO activity via unknown mechanisms [64,65]. Once these mechanisms are solved, the development of defensin-like molecules could constitute promising therapeutic tools against L. monocytogenes and other CDC-producing pathogens [66]. ...
... A similar enzymatic process was recently proposed to exist in neutrophils in which the matrix metalloproteinase-8, stored in secondary granules, degrades LLO [109]. Also, the antimicrobial peptides, α-defensins, stored at high concentrations in neutrophil primary granules, inhibit LLO release from the bacterium in addition to blocking its activity [64,65]. Paradoxically, during L. monocytogenes phagocytosis by neutrophils, LLO triggers its own inactivation by stimulating the exocytosis of secondary and primary granules [109]. ...
Article
The cholesterol-dependent cytolysins (CDCs) are a large family of pore-forming toxins that are produced by numerous Gram-positive bacterial pathogens. These toxins are released in the extracellular environment as water-soluble monomers or dimers that bind to cholesterol-rich membranes and assemble into large pore complexes. Depending upon their concentration, the nature of the host cell and membrane (cytoplasmic or intracellular) they target, the CDCs can elicit many different cellular responses. Among the CDCs, listeriolysin O (LLO), which is a major virulence factor of the facultative intracellular pathogen Listeria monocytogenes, is involved in several stages of the intracellular lifecycle of the bacterium and displays unique characteristics. It has long been known that following L. monocytogenes internalization into host cells, LLO disrupts the internalization vacuole, enabling the bacterium to replicate into the host cell cytosol. LLO is then used by cytosolic bacteria to spread from cell to cell, avoiding bacterial exposure to the extracellular environment. Although LLO is continuously produced during the intracellular lifecycle of L. monocytogenes, several processes limit its toxicity to ensure the survival of infected cells. It was previously thought that LLO activity was limited to mediating vacuolar escape during bacterial entry and cell to cell spreading. This concept has been challenged by compelling evidence suggesting that LLO secreted by extracellular L. monocytogenes perforates the host cell plasma membrane, triggering important host cell responses. This chapter provides an overview of the well-established intracellular activity of LLO and the multiple roles attributed to LLO secreted by extracellular L. monocytogenes.
... The mechanism by which rHNP-1 reduces amastigote load in macrophages is unknown, but may be a direct effect of rHNP-1 on parasites. In this respect, Arnett et al. have shown that HNP-1 is taken up by bacteria-infected macrophages and reduces bacterial load [31]. ...
... HNP-1 has been shown to act as a chemotactic factor for both macrophages and T lymphocytes [11] and HNP-1 treatment enhances leukocyte accumulation at the site of infection in K. pneumonia-infected mice [30] and provokes TNF-a release from lymphocytes [32]. Further, HNP-1 treatment of macrophages has been shown to inhibit proliferation of the intracellular bacteria [31]. Another immunomodulatory molecule is CpG motif. ...
Article
Full-text available
The toxicity of available drugs for treatment of leishmaniasis, coupled with emerging drug resistance, make it urgent to find new therapies. Antimicrobial peptides (AMPs) have a strong broad-spectrum antimicrobial activity with distinctive modes of action and are considered as promising therapeutic agents. The defensins, members of the large family of AMPs, are immunomodulatory molecules and important components of innate immune system. Human neutrophil peptide-1 (HNP-1), which is produced by neutrophils, is one of the most potent defensins. In this study, we described anti-parasitic activity of recombinant HNP-1 (rHNP-1) against Leishmania major promastigotes and amastigotes. Furthermore, we evaluated the immunomodulatory effect of rHNP-1 on parasite-infected neutrophils and how neutrophil apoptosis was affected. Our result showed that neutrophils isolated from healthy individuals were significantly delayed in the onset of apoptosis following rHNP-1 treatment. Moreover, there was a noteworthy increase in dying cells in rHNP-1- and/or CpG-treated neutrophils in comparison with untreated cells. There is a considerable increase in TNF-α production from rHNP-1-treated neutrophils and decreased level of TGF-β concentration, a response that should potentiate the immune system against parasite invasion. In addition, by using real-time polymerase chain reaction (real-time PCR), we showed that in vitro infectivity of Leishmania into neutrophils is significantly reduced following rHNP-1 treatment compared to untreated cells.
... Retrocyclin is a circular θ-defensin peptide which has been artificially humanised and was previously reported to have broad antimicrobial activity [89][90][91][92]. Recent research has shown that the novel θ-defensin retrocyclin-101 (RC-101) has inhibitory activity against JEV infection. ...
Article
Full-text available
Japanese encephalitis virus (JEV) belongs to the Flaviviridae family and is a representative mosquito-borne flavivirus responsible for acute encephalitis and meningitis in humans. Despite the availability of vaccines, JEV remains a major public health threat with the potential to spread globally. According to the World Health Organization (WHO), there are an estimated 69,000 cases of JE each year, and this figure is probably an underestimate. The majority of JE victims are children in endemic areas, and almost half of the surviving patients have motor or cognitive sequelae. Thus, the absence of a clinically approved drug for the treatment of JE defines an urgent medical need. Recently, several promising and potential drug candidates were reported through drug repurposing studies, high-throughput drug library screening, and de novo design. This review focuses on the historical aspects of JEV, the biology of JEV replication, targets for therapeutic strategies, a target product profile, and drug development initiatives.
... All donors provided informed, written consent. BMDMs were obtained from 89-wk-old female C57BL/6 mice (The Jackson Laboratory) (26). ...
Article
AIM2 (absent in melanoma 2), an inflammasome component, mediates IL-1β release in murine macrophages and cell lines. AIM2 and IL-1β contribute to murine control of Mycobacterium tuberculosis (M.tb) infection, but AIM2’s impact in human macrophages, the primary niche for M.tb, remains unclear. We show that M.tb, Mycobacterium bovis bacillus Calmette–Guérin (BCG), and M. smegmatis induce AIM2 expression in primary human macrophages. M.tb-induced AIM2 expression is peroxisome proliferator-activated receptor γ (PPARγ)–dependent and M.tb ESX-1–independent, whereas BCG- and M. smegmatis–induced AIM2 expression is PPARγ-independent. PPARγ and NLRP3, but not AIM2, are important for IL-1β release in response to M.tb, and NLRP3 colocalizes with M.tb. This is in contrast to the role for AIM2 in inflammasome activation in mice and peritoneal macrophages. Altogether, we show that mycobacteria induce AIM2 expression in primary human macrophages, but AIM2 does not contribute to IL-1β release during M.tb infection, providing further evidence that AIM2 expression and function are regulated in a cell- and/or species-specific manner.
... Our finding that α-defensin-1 is taken-up into all tested cell lines serves our aim of using it as a therapeutic drug reaching intracellular targets. This finding is in line with previous studies of other groups, which found that α-defensin-1 accumulates intracellularly in human cervical CaSki cells and inhibits human simplex virus, also post-entry [55] and is also taken up into bone marrow-derived macrophages [56]. While α-defensin-5, found in the intestine, does not physiologically come into contact with PT, which mainly acts in the respiratory tract, it might have therapeutic implications as an inhibitor of PT uptake into cells. ...
Article
Full-text available
Whooping cough is a severe childhood disease, caused by the bacterium Bordetella pertussis, which releases pertussis toxin (PT) as a major virulence factor. Previously, we identified the human antimicrobial peptides α-defensin-1 and -5 as inhibitors of PT and demonstrated their capacity to inhibit the activity of the PT enzyme subunit PTS1. Here, the underlying mechanism of toxin inhibition was investigated in more detail, which is essential for developing the therapeutic potential of these peptides. Flow cytometry and immunocytochemistry revealed that α-defensin-5 strongly reduced PT binding to, and uptake into cells, whereas α-defensin-1 caused only a mild reduction. Conversely, α-defensin-1, but not α-defensin-5 was taken up into different cell lines and interacted with PTS1 inside cells, based on proximity ligation assay. In-silico modeling revealed specific interaction interfaces for α-defensin-1 with PTS1 and vice versa, unlike α-defensin-5. Dot blot experiments showed that α-defensin-1 binds to PTS1 and even stronger to its substrate protein Gαi in vitro. NADase activity of PTS1 in vitro was not inhibited by α-defensin-1 in the absence of Gαi. Taken together, these results suggest that α-defensin-1 inhibits PT mainly by inhibiting enzyme activity of PTS1, whereas α-defensin-5 mainly inhibits cellular uptake of PT. These findings will pave the way for optimization of α-defensins as novel therapeutics against whooping cough.
... The design and synthesis of chimeric CPP-AMP peptides with a cleavable link also comprise an attractive and efficient mechanistic option to kill intracellular pathogens [41,42]. Another exciting mechanism to kill intracellular pathogens is via a cooperative action of the AMPS α-defensins and humanized θ-defensin with macrophages that entrapped the bacteria in the phagosome, where they are annihilate [43]. The following sections present examples of these cell-penetrating antimicrobial peptides with antibacterial, antiviral, and antiparasitic activities. ...
Article
Full-text available
Cell-penetrating peptides (CPPs) are natural or engineered peptide sequences with the intrinsic ability to internalize into a diversity of cell types and simultaneously transport hydrophilic molecules and nanomaterials, of which the cellular uptake is often limited. In addition to this primordial activity of cell penetration without membrane disruption, multivalent antimicrobial activity accompanies some CPPs. Antimicrobial peptides (AMPs) with cell-penetrability exert their effect intracellularly, and they are of great interest. CPPs with antimicrobial activity (CPAPs) comprise a particular class of bioactive peptides that arise as promising agents against difficult-to-treat intracellular infections. This short review aims to present the antibacterial, antiparasitic, and antiviral effects of various cell-penetrating antimicrobial peptides currently documented. Examples include the antimicrobial effects of different CPAPs against bacteria that can propagate intracellularly, like Staphylococcus sp., Streptococcus sp., Chlamydia trachomatis, Escherichia coli, Mycobacterium sp., Listeria sp., Salmonella sp. among others. CPAPs with antiviral effects that interfere with the intracellular replication of HIV, hepatitis B, HPV, and herpes virus. Additionally, CPAPs with activity against protozoa of the genera Leishmania, Trypanosoma, and Plasmodium, the etiological agents of Leishmaniasis, Chagas’ Disease, and Malaria, respectively. The information provided in this review emphasizes the potential of multivalent CPAPs, with anti-infective properties for application against various intracellular infections. So far, CPAPs bear a promise of druggability for the translational medical use of CPPs alone or in combination with chemotherapeutics. Moreover, CPAPs could be an exciting alternative for pharmaceutical design and treating intracellular infectious diseases.
... hBD-3 can also stimulate monocytes and macrophages for secretion of a variety of chemokines including macrophage-derived chemokine, the angiogenic factor and vascular endothelial growth factor [117]. Although not expressed by macrophages, defensins can contribute to the antibacterial function of the phagocytes by inhibiting phagosomal escape and intracellular multiplication of Listeria monocytogenes and Mycobacterium tuberculosis [118,119]. Of note, HNP1 inhibits macrophage-driven inflammation in the aftermath of Salmonella infection, ensuring both pathogen clearance and inflammation resolution [120]. ...
Article
Full-text available
Defensins are a family of cationic antimicrobial peptides active against a broad range of infectious microbes including bacteria, viruses and fungi, playing important roles as innate effectors and immune modulators in immunological control of microbial infection. Their antibacterial properties and unique mechanisms of action have garnered considerable interest in developing defensins into a novel class of natural antibiotic peptides to fend off pathogenic infection by bacteria, particularly those resistant to conventional antibiotics. However, serious pharmacological and technical obstacles, some of which are unique to defensins and others are common to peptide drugs in general, have hindered the development and clinical translation of defensins as anti-infective therapeutics. To overcome them, several technologies have been developed, aiming for improved functionality, prolonged circulation time, enhanced proteolytic stability and bioavailability, and efficient and controlled delivery and release of defensins to the site of infection. Additional challenges include the alleviation of potential toxicity of defensins and their cost-effective manufacturing. In this review, we briefly introduce defensin biology, focus on various transforming strategies and practical techniques developed for defensins and their derivatives as antibacterial therapeutics, and conclude with a summation of future challenges and possible solutions.
... Retrocyclin (RC) is an artificially humanized u -defensin that has been reported to possess broad antimicrobial activity (16)(17)(18)(19)(20)(21). RC-101 has the sequence GICRCICGKGICRCICGR and is an analogue of RC-1 (GICRCICGRGICRCICGR). ...
Article
Full-text available
Currently, there are no approved drugs for the treatment of flavivirus infection. Accordingly, we tested the inhibitory effects of the novel θ-defensin retrocyclin-101 (RC-101) against flavivirus infection, and investigated the mechanism underlying the potential inhibitory effects. First, RC-101 robustly inhibited both Japanese encephalitis virus (JEV) and Zika virus (ZIKV) infections. RC-101 exerted inhibitory effects on the entry and replication stages. Results also indicated that the non-structural protein NS2B-NS3 serine protease might serve as a potential viral target. Further, RC-101 inhibited protease activity at the micromolar level. We also demonstrated that with respect to the glycoprotein E protein of flavivirus, the DE loop of domain III, which is the receptor-binding domain of the E protein, might serve as another viral target of RC-101. Moreover, a JEV DE mutant exhibited resistance to RC-101, which was associated with deceased binding affinity of RC-101 to DIII. These findings provide a basis for the development of RC-101 as a potential candidate for the treatment of flavivirus infection. Importance Retrocyclin is an artificially humanized circular θ-defensin peptide, containing 18 residues previously reported to possess broad antimicrobial activity. In this study, we found that retrocyclin-101 inhibited flavivirus (ZIKV and JEV) infections. Retrocyclin-101 inhibited NS2B-NS3 serine protease activity, suggesting that the catalytic triad of the protease is the target. Moreover, retrocyclin-101 bound to the DE loop of the E protein of flavivirus, which prevented its entry.
... HNP1-3 secreted by infiltrating neutrophils in Staphylococcus aureus infection induce TNF-α and IFN-γ release from macrophages, which, in turn, increase phagocytosis of pathogens (163) neck and head oral by macrophages -an essential step in bacterial clearance (84,85). HNP1 also inhibits phagosomal escape and intracellular multiplication of Listeria monocytogenes and Mycobacterium tuberculosis in macrophages (86,87), suggesting that the defensin, although not being expressed by macrophages, contributes to their antimicrobial function. Notably, HNP1 acts in the aftermath of Salmonella infection as a "molecular brake" on macrophage-driven inflammation by preventing protein translation to ensure both pathogen clearance and the resolution of inflammation with minimal bystander tissue damage (88). ...
Article
Full-text available
Defensins are a major family of host defense peptides expressed predominantly in neutrophils and epithelial cells. Their broad antimicrobial activities and multifaceted immunomodulatory functions have been extensively studied, cementing their role in innate immunity as a core host-protective component against bacterial, viral and fungal infections. More recent studies, however, paint defensins in a bad light such that they are “alleged” to promote viral and bacterial infections in certain biological settings. This mini review summarizes the latest findings on the potential pathogenic properties of defensins against the backdrop of their protective roles in antiviral and antibacterial immunity. Further, a succinct description of both tumor-proliferative and -suppressive activities of defensins is also given to highlight their functional and mechanistic complexity in antitumor immunity. We posit that given an enabling environment defensins, widely heralded as the “Swiss army knife,” can function as a “double−edged sword” in host immunity.
... Nevertheless, more sophisticated antimicrobial mechanisms of defensins were deciphered in recent studies, including inhibition of bacterial cell wall synthesis, interference of membrane function, neutralization of bacterial toxins, and formation of nanonet "traps" to restrain pathogens in host intestine (7,9,10,52,53). Besides these direct antimicrobial effects, synergistic interactions between defensins and immune cells, especially with macrophages, are believed to provide assistance for the control of bacterial infection, including Mycobacterium tuberculosis, Legionella pneumophila, Staphylococcus aureus, and Listeria monocytogenes, through boosting the phagocytosis and further inhibiting phagosomal escape and intracellular multiplication (29,(31)(32)(33)(34)(35)54). A recent study revealed HNP1 functions as a "molecular brake" on macrophagedriven inflammation caused by Salmonella by preventing protein translation, ensuring both pathogen clearance and the resolution of inflammation with minimal bystander tissue damage (55). ...
Article
Full-text available
Human α-defensins are 3-5 KDa, disulfide-bridged peptides with a multiplitude of anti-microbial activities and immunomodulatory functions. Recent studies show that human enteric α-defensin 5 (HD5), a host defense peptide important for intestinal homeostasis and innate immunity, aids the highly infectious enteropathogen Shigella to breach the intestinal epithelium in vitro and in vivo . Whether or not and how HD5 influences Shigella infection of resident macrophages following its invasion of the epithelial epithelium remains poorly understood. Here we report that HD5 greatly promoted phagocytosis of Shigella by macrophages by targeting the bacteria to enhance bacterium-to-cell contacts in a structure- and sequence-dependent fashion. Subsequent intracellular multiplication of phagocytosed Shigella led to massive necrotic cell death and release of the bacteria. HD5-promoted phagocytosis of Shigella was independent of the status of the type 3 secretion system. Further, HD5 neither inhibited nor enhanced phagosomal escape of Shigella . Collectively, these findings confirm a potential pathogenic role of HD5 in Shigella infection of not only epithelial cells but also macrophages, illuminating how an enteropathogen exploits a host protective factor for virulence and infection.
... In summary, the present results have revealed the potentiality of human secretory RNases to work as antimicrobial agents in the fight against tuberculosis, extendable to other intracellular infectious bacterial diseases that are difficult to eradicate (78). AMPs that display a multifaceted mechanism of action (12,16,(79)(80)(81)(82)(83)(84)(85), like human antimicrobial RNases, deserve a special attention for their pharmacological potential. Our ongoing interdisciplinary investigation warrants the understanding of RNases' antimicrobial mechanism of action and can pave the design of novel anti-infective therapies. ...
Article
Full-text available
The development of novel treatment against tuberculosis is a priority global health challenge. Antimicrobial proteins and peptides offer a multifaceted mechanism suitable to fight bacterial resistance. Within the RNaseA superfamily there is a group of highly cationic proteins secreted by innate immune cells with anti-infective and immune-regulatory properties. In this work, we have tested the human canonical members of the RNase family using a spot-culture growth inhibition assay based mycobacteria-infected macrophage model for evaluating their anti-tubercular properties. Out of the seven tested recombinant human RNases, we have identified two members, RNase3 and RNase6, which were highly effective against Mycobacterium aurum extra-and intracellularly and induced an autophagy process. We observed the proteins internalization within macrophages and their capacity to eradicate the intracellular mycobacterial infection at a low micro-molar range. Contribution of the enzymatic activity was discarded by site-directed mutagenesis at the RNase catalytic site. The protein induction of autophagy was analyzed by RT-qPCR, western blot, immunofluorescence, and electron microscopy. Specific blockage of auto-phagosome formation and maturation reduced the protein's ability to eradicate the infection. In addition, we found that the M. aurum infection of human THP1 macrophages modulates the expression of endogenous RNase3 and RNase6, suggesting a function in vivo. Overall, our data anticipate a biological role for human antimicrobial RNases in host response to mycobacterial infections and set the basis for the design of novel anti-tubercular drugs.
... This amino acid is missing in HNP-2 peptide, and HNP-2 is thought to be a proteolytic product of HNP-1 and HNP-3 (16,17). In addition to their broad repertoire of antimicrobial activities (18,19), HNP1-3 exert multiple immunomodulatory effects (20)(21)(22), as well as early release alarmin activity to initiate the host response upon microbial invasion or tissue injury/damage (23). Studies in patients have documented that the levels of HNP1-3 in various body fluids (e.g., blood, bronchoalveolar lavage fluid, and sputum) are greatly increased during sepsis (24). ...
Article
Full-text available
Significance Genetic copy number variations (CNVs) are the most common mechanism of structural genetic diversity, playing a key role in human health and common diseases. However, because of a lack of relevant experimental models, little is known about how rare CNVs contribute to the risk of complex phenotypes. We show that transgenic mice with high copy number of DEFA1/DEFA3 [encoding human neutrophil peptides 1–3 (HNP1–3)] suffer from more severe sepsis because of more extensive endothelial barrier dysfunction and endothelial cell pyroptosis. Functional blockade of HNP1–3 rescues the mice from lethal sepsis. These findings exemplify how CNVs modulate disease development in appropriate animal models and explore a paradigm for the precision treatment of sepsis tailored by individual genetic information.
... No degradative pathway for their removal from tissue has been identified. Another possible explanation for this decrement is the ingestion of HNPs 1-3 by peripheral blood mononuclear cells, for previous studies have shown that these cells can capture defensins in various ways after a much mount of HNPs suddenly released into the circulation [29,36]. In addition, the expression of α-defensins significantly correlated with the proportion and number of airway neutrophils [19]. ...
Article
Full-text available
Abstract Background Early onset of lung injury is considerable common after cardiac surgery and is associated with increasing in morbidity and mortality, but current clinical predictors for the occurrence of this complication always have limited positive warning value. This study aimed to evaluate whether elevated plasma levels of human neutrophil peptides (HNPs) 1–3 herald impaired lung function in infants and young children after cardiac surgery necessitating cardiopulmonary bypass (CPB). Methods Consecutive children younger than 3 years old who underwent cardiac surgery were prospectively enrolled. Plasma concentrations of HNPs 1–3 and inflammatory cytokines were measured before, and immediately after CPB, as well as at 1 h, 12 h, and 24 h after CPB. Results Thirty patients were enrolled, 18 (60%) of whom were infants. Plasma levels of HNPs 1–3 and the pro-inflammatory cytokine interleukin-6 (IL-6) significantly increased immediately after CPB (P
... Defensins are small, cysteine-rich, cationic peptides that primarily serve as innate immune defense mechanisms against infectious microorganisms. Unlike the aand b-defensins, u-defensins are circular, formed by head-to-tail ligation of 2 truncated a-defensin-like precursor peptides and possess broad antimicrobial activity, targeting bacteria, viruses, and fungi [28][29][30][31][32]. All 6 human u-defensin (DEFT) pseudogenes contain a premature stop codon, preventing translation [33,34]. ...
Article
Despite widespread use of annual influenza vaccines, seasonal influenza-associated deaths number in the thousands each year, in part because of exacerbating bacterial superinfections. Therefore, discovering additional therapeutic options would be a valuable aid to public health. Recently, TLR4 inhibition has emerged as a possible mechanism for protection against influenza-associated lethality and acute lung injury. Based on recent data showing that rhesus macaque θ-defensins could inhibit TLR4-dependent gene expression, we tested the hypothesis that a novel θ-defensin, retrocyclin (RC)-101, could disrupt TLR4-dependent signaling and protect against viral infection. In this study, RC-101, a variant of the humanized θ-defensin RC-1, blocked TLR4-mediated gene expression in mouse and human macrophages in response to LPS, targeting both MyD88- and TRIF-dependent pathways. In a cell-free assay, RC-101 neutralized the biologic activity of LPS at doses ranging from 0.5 to 50 EU/ml, consistent with data showing that RC-101 binds biotinylated LPS. The action of RC-101 was not limited to the TLR4 pathway because RC-101 treatment of macrophages also inhibited gene expression in response to a TLR2 agonist, Pam3CSK4, but failed to bind that biotinylated agonist. Mouse macrophages infected in vitro with mouse-adapted A/PR/8/34 influenza A virus (PR8) also produced lower levels of proinflammatory cytokine gene products in a TLR4-independent fashion when treated with RC-101. Finally, RC-101 decreased both the lethality and clinical severity associated with PR8 infection in mice. Cumulatively, our data demonstrate that RC-101 exhibits therapeutic potential for the mitigation of influenza-related morbidity and mortality, potentially acting through TLR-dependent and TLR-independent mechanisms.
... The phagolysosome is typically acidic at approximately pH 5.0, which inhibits the filamentation of Candida albicans. The phagolysosome of neutrophils also contains degradative enzymes and inhibitory proteins (such as elastase, lactoferrin and myeloperoxidase) and antimicrobial peptides (including, defensins and cathelicidins such as LL37), which have been shown in antibacterial immunity to be able to be transferred to macrophages 131 Figure 4 | Fungal survival strategies. a | The toxic nature of the phagolysosome induces a wide range of survival mechanisms that mitigate the potency of oxidative, nitrosative, pH and other stresses that act to kill fungal pathogens. ...
Article
Full-text available
The surveillance and elimination of fungal pathogens rely heavily on the sentinel behaviour of phagocytic cells of the innate immune system, especially macrophages and neutrophils. The efficiency by which these cells recognize, uptake and kill fungal pathogens depends on the size, shape and composition of the fungal cells and the success or failure of various fungal mechanisms of immune evasion. In this Review, we describe how fungi, particularly Candida albicans, interact with phagocytic cells and discuss the many factors that contribute to fungal immune evasion and prevent host elimination of these pathogenic microorganisms.
... Many HDPs including cathelicidins, defensins and hepcidin, have been demonstrated to protect against bacterial, viral and parasitic infections (15,(29)(30)(31)(32)(33)(34). Several studies provide evidence to correlate the expression of HDPs with susceptibility or resistance to bacterial infections (35)(36)(37). ...
Article
Natural cationic host defence (antimicrobial) peptides are widely distributed gene encoded molecules with diverse structures. There are more than 1200 natural Host Defence Peptides (HDPs) described to date. Due to the multifunctional roles defined for such peptides there is a keen interest in the potential therapeutic applications of HDPs and their synthetic mimics, Antimicrobial peptides and Innate Defence Regulator (IDR) peptides. These peptides constitute two broad classes of potential therapeutics; (i) with direct antimicrobial and/or anti-biofilm activity, and (ii) with immune-modulating and/or anti-inflammatory activity. Exploiting the immunomodulatory functions of these peptides represents a new therapeutic approach for resolution of infections and inflammatory disorders.
... Finally, similar to αand β-defensins, retrocyclins were shown to inhibit several unrelated bacterial toxins, (e.g. Bacillus anthracis toxin [14], Gardnerella vaginalis toxin [15], and Listeria monocytogenes listeriolysin O (LLO) [16]), but the mechanisms of selective inhibition of toxins (without affecting host proteins) remained enigmatic. ...
Article
Full-text available
Defensins are a class of immune peptides with a broad range of activities against bacterial, fungal, and viral pathogens. Besides exerting direct antimicrobial activity via disorganization of bacterial membranes, defensins are also able to neutralize various unrelated bacterial toxins. Recently we have demonstrated that in the case of human α- and β-defensins this later ability is achieved through exploiting toxins' marginal thermodynamic stability, i.e. defensins act as molecular anti-chaperones unfolding toxin molecules and exposing their hydrophobic regions and thus promoting toxin precipitation and inactivation [Kudryashova, Quintyn, Seveau, Lu, Wysocki, Kudryashov (2014) Immunity 41, 709-721]. Retrocyclins are humanized synthetic θ-defensin peptides that possess unique cyclic structure, differentiating them from α- and β-defensins. Importantly, retrocyclins are more potent against some bacterial and viral pathogens and more stable than their linear counterparts. However, the mechanism of bacterial toxin inactivation by retrocyclins is not known. In the present study we demonstrate that retrocyclins facilitate unfolding of bacterial toxins. Using differential scanning fluorimetry, limited proteolysis, and collisional quenching of internal tryptophan fluorescence, we show that hydrophobic regions of toxins normally buried in the molecule interior become more exposed to solvents and accessible to proteolytic cleavage in the presence of retrocyclins. The retrocyclin-induced unfolding of toxins led to their precipitation and abrogated activity. Toxin inactivation by retrocyclins was strongly diminished under reducing conditions, but preserved at physiological salt and serum concentrations. Therefore, despite significant structural diversity, α-, β-, and θ-defensins employ similar mechanisms of toxin inactivation, which may be shared by antimicrobial peptides from other families.
... It is tempting to speculate that the proposed vulnerability of marginally stable proteins might extend well beyond bacterial toxins and include secretion machineries of Gram-positive bacteria (Arnett et al., 2011;Vega and Caparon, 2012), as well as numerous capsid and noncapsid viral proteins (Furci et al., 2007;Gounder et al., 2012). Many viral proteins display loosely packed cores (a hallmark of thermodynamic instability) that provide evolutionary advantage by conferring high interactive promiscuity and high mutational adaptability (Tokuriki et al., 2009;Wylie and Shakhnovich, 2011). ...
Article
Full-text available
Defensins are short cationic, amphiphilic, cysteine-rich peptides that constitute the front-line immune defense against various pathogens. In addition to exerting direct antibacterial activities, defensins inactivate several classes of unrelated bacterial exotoxins. To date, no coherent mechanism has been proposed to explain defensins’ enigmatic efficiency toward various toxins. In this study, we showed that binding of neutrophil α-defensin HNP1 to affected bacterial toxins caused their local unfolding, potentiated their thermal melting and precipitation, exposed new regions for proteolysis, and increased susceptibility to collisional quenchers without causing similar effects on tested mammalian structural and enzymatic proteins. Enteric α-defensin HD5 and β-defensin hBD2 shared similar toxin-unfolding effects with HNP1, albeit to different degrees. We propose that protein susceptibility to inactivation by defensins is contingent to their thermolability and conformational plasticity and that defensin-induced unfolding is a key element in the general mechanism of toxin inactivation by human defensins.
... 51 Moreover, modified θ -defensin (RC-1) inhibited the growth of Listeria monocytogenes in macrophages more remarkably than α -defensin HNP-1. 52 The results suggest that θ -defensin is applicable to the human body. ...
Article
Full-text available
Mucosal surface of the intestinal tract is continuously exposed to a large number of microorganisms. To manage the substantial microbial exposure, epithelial surfaces produce a diverse arsenal of antimicrobial proteins (AMPs) that directly kill or inhibit the growth of microorganisms. Thus, AMPs are important components of innate immunity in the gut mucosa. They are frequently expressed in response to colonic inflammation and infection. Expression of many AMPs, including human β-defensin 2-4 and cathelicidin, is induced in response to invasion of pathogens or enteric microbiota into the mucosal barrier. In contrast, some AMPs, including human α-defensin 5-6 and human β-defensin 1, are constitutively expressed without microbial contact or invasion. In addition, specific AMPs are reported to be associated with inflammatory bowel disease (IBD) due to altered expression of AMPs or development of autoantibodies against AMPs. The advanced knowledge for AMPs expression in IBD can lead to its potential use as biomarkers for disease activity. Although the administration of exogenous AMPs as therapeutic strategies against IBD is still at an early stage of development, augmented induction of endogenous AMPs may be another interesting future research direction for the protective and therapeutic purposes. This review discusses new advances in our understanding of how intestinal AMPs protect against pathogens and contribute to pathophysiology of IBD.
... [91] Furthermore, retrocyclin-1 is active against L. monocytogenes and inhibits listeriolysin O, a pore-forming toxin that enables bacteria to escape from phagosomes. [92] Figure 5. Chemical synthesis of q-defensins involves three stages: assembly of the peptide chain on resin; cleavage from the resin; and cyclization and oxidation to form the circular backbone and three disulfide bonds. ...
Article
Cyclic peptides are found in a diverse range of organisms and are characterized by their stability and role in defense. Why is only one class of cyclic peptides found in mammals? Possibly we have not looked hard enough for them, or the technologies needed to identify them are not fully developed. We also do not yet understand their intriguing biosynthesis from two separate gene products. Addressing these challenges will require the application of chemical tools and insights from other classes of cyclic peptides. Herein, we highlight recent developments in the characterization of theta defensins and describe the important role that chemistry has played in delineating their modes of action. Furthermore, we emphasize the potential of theta defensins as antimicrobial agents and scaffolds for peptide drug design.
... CHDPs are known to confer protection against bacterial, viral and parasitic infections, and modulate immune responses in the host. [1][2][3][4][5][6][7][8][9][10] Research in the last two decades has demonstrated a wide range of immune functions mediated by CHDPs that influence both innate and adaptive immunity (reviewed in refs 11-13). The two best characterized families of CHDPs in mammals are cathelicidins and defensins. ...
Article
Full-text available
Human cathelicidin LL-37 protects against infections and endotoxin-induced inflammation. In a recent study we have shown that IG-19, a LL-37-derived peptide, protects in a murine model of arthritis. Cytokine IL-32 is elevated and directly associated with the disease severity of inflammatory arthritis. Therefore, in this study we examined the effects of LL-37 and IG-19 on IL-32-induced responses in human peripheral blood-derived mononuclear cells (PBMC) and macrophages. We showed that CD14+ monocytes are the primary cells that produce pro-inflammatory TNF-α following stimulation of PBMC with IL-32. We demonstrated that LL-37 and IG-19 significantly suppress IL-32-induced production of pro-inflammatory cytokines e.g. TNF-α and IL-1β, without altering chemokine production. In contrast, LL-37 and IG-19 enhance the production of the anti-inflammatory cytokine IL-1RA. Further mechanistic studies revealed that LL-37 and IG-19 suppress IL-32-mediated phosphorylation of Fyn (Y420) Src kinase. In contrast, IL-32-mediated phosphorylation of AKT-1 (T308) and MKP-1 (S359) is not suppressed by the peptides. LL-37 and IG-19 alone induce the phosphorylation of MKP-1 (S359), which is a known negative regulator of inflammation. Furthermore, the peptides induce the activity of p44/42 MAPK, which is known to phosphorylate MKP-1 (S359). This is the first study to demonstrate the regulation of IL-32-induced inflammation by LL-37 and its derivative peptide IG-19. The mechanistic results from this study suggest that regulation of immune-mediated inflammation by these peptides may be controlled by the dual phosphatase MKP-1. We speculate that LL-37 and its derivatives may contribute to the control of immune-mediated inflammatory diseases. This article is protected by copyright. All rights reserved.
... Cells were used at passage 2-6 and prepared prior to infection as described. 54,55 Glass coverslips were coated with rat tail collagen prior to seeding Caco-2 cells at 1.0 × 10 5 cells/well in 24-well cell culture plates. Overnight cultures of the bacterial strains were diluted to an OD 600 of 0.1 in 5 mL of BHI and grown to mid-exponential phase (OD 600 of 0.8). ...
Article
Listeria monocytogenes is an intracellular, foodborne gastrointestinal pathogen that is primarily responsible for causing listeriosis or food poisoning in otherwise healthy individuals. Infections that arise during pregnancy or within immune compromised individuals are much more serious resulting in the risk of fetal termination or fetal fatality postpartum in the former and septicemia or meningitis with a 20% fatality rate in the latter. While the roles of internalin proteins and listeriolysin-O in the infection process are well characterized, the specific roles of lysine-modified phospholipids in the membrane of L. monocytogenes are not. Investigation into the lipid bilayer composition of L. monocytogenes indicated that the overall proportions of lipids, including lysylcardiolipin and lysylphosphatidylglycerol (LysPG), vary with growth temperature and growth phase. In addition, we demonstrate that LysPG formation is essential for L. monocytogenes survival in the presence of increased osmolytic stress but has no effect on bacterial adherence, invasion or survival in the presence of physiologically relevant concentrations of human neutrophil peptide (HNP-1). In the absence of LysPG synthesis, L. monocytogenes unexpectedly retained flagellum-mediated motility at 37 °C. Taken together, these findings show that LysPG formation in L. monocytogenes has broader functions in virulence and survival beyond its known role in the modification of membrane potential previously observed in other bacteria.
... This implies a potential role for H␤D2 in the observed resistance to intracellular infection. Accumulation of intracellular defensins has been demonstrated to inhibit replication of the obligate intracellular pathogen Listeria monocytogenes in macrophages (69), and a similar mechanistic link between H␤D2 and resistance to intracellular Salmonella may exist in milk HA-treated intestinal epithelium. Although questions regarding the mechanism of milk HA-mediated Salmonella infection resistance remain, our results suggest that milk HA may contribute to the reduced incidence of enteric Salmonella infection associated with breast-feeding in human infants (7). ...
Article
Full-text available
Background Human milk contains hyaluronan (HA). Results: Milk HA concentration is highest immediately after delivery. Treatment of epithelium with physiologic levels of milk-derived HA increases intracellular expression of β-defensin (in vitro and in vivo) and resistance to Salmonella. Conclusion: Milk HA enhances functional antimicrobial defense mechanisms of the intestinal epithelium. Significance: Milk HA may be a mediator of maternal protection of newborns. Breast-feeding is associated with enhanced protection from gastrointestinal disease in infants, mediated in part by an array of bioactive glycan components in milk that act through molecular mechanisms to inhibit enteric pathogen infection. Human milk contains hyaluronan (HA), a glycosaminoglycan polymer found in virtually all mammalian tissues. We have shown that synthetic HA of a specific size range promotes expression of antimicrobial peptides in intestinal epithelium. We hypothesize that hyaluronan from human milk also enhances innate antimicrobial defense. Here we define the concentration of HA in human milk during the first 6 months postpartum. Importantly, HA isolated from milk has a biological function. Treatment of HT-29 colonic epithelial cells with human milk HA at physiologic concentrations results in time- and dose-dependent induction of the antimicrobial peptide human β-defensin 2 and is abrogated by digestion of milk HA with a specific hyaluronidase. Milk HA induction of human β-defensin 2 expression is also reduced in the presence of a CD44-blocking antibody and is associated with a specific increase in ERK1/2 phosphorylation, suggesting a role for the HA receptor CD44. Furthermore, oral administration of human milk-derived HA to adult, wild-type mice results in induction of the murine Hβ D2 ortholog in intestinal mucosa and is dependent upon both TLR4 and CD44 in vivo. Finally, treatment of cultured colonic epithelial cells with human milk HA enhances resistance to infection by the enteric pathogen Salmonella typhimurium. Together, our observations suggest that maternally provided HA stimulates protective antimicrobial defense in the newborn.
... Various models are proposed for the microbicidal activities of antimicrobial cationic peptides, which include interaction with the negatively charged membrane components of microbes resulting in pore formation, induction of non-specific membrane permeabilization, binding to intracellular targets and disruption of bacterial biofilms [3,4] . Natural cationic antimicrobial peptides can indeed protect against a wide range of infections, including bacterial, viral and parasitic [5][6][7][8] . However, it is now appre-ciated that the direct microbicidal activity of certain cationic antimicrobial peptides, e.g. ...
Article
Full-text available
Host defence peptides (HDPs) are innate immune effector molecules found in diverse species. HDPs exhibit a wide range of functions ranging from direct antimicrobial properties to immunomodulatory effects. Research in the last decade has demonstrated that HDPs are critical effectors of both innate and adaptive immunity. Various studies have hypothesized that the antimicrobial property of certain HDPs may be largely due to their immunomodulatory functions. Mechanistic studies revealed that the role of HDPs in immunity is very complex and involves various receptors, signalling pathways and transcription factors. This review will focus on the multiple functions of HDPs in immunity and inflammation, with special reference to cathelicidins, e.g. LL-37, certain defensins and novel synthetic innate defence regulator peptides. We also discuss emerging concepts of specific HDPs in immune-mediated inflammatory diseases, including the potential use of cationic peptides as therapeutics for immune-mediated inflammatory disorders.
Article
Tuberculosis (TB) accounts for 1.6 million deaths annually and over 25% of deaths due to antimicrobial resistance. Mycobacterium tuberculosis (M.tb) drives MCL-1 expression (family member of anti-apoptotic BCL-2 proteins) to limit apoptosis and grow intracellularly in human macrophages. The feasibility of re-purposing specific MCL-1 and BCL-2 inhibitors to limit M.tb growth, using inhibitors that are in clinical trials and FDA-approved for cancer treatment has not be tested previously. We show that specifically inhibiting MCL-1 and BCL-2 induces apoptosis of M.tb-infected macrophages, and markedly reduces M.tb growth in human and murine macrophages, and in a pre-clinical model of human granulomas. MCL-1 and BCL-2 inhibitors limit growth of drug resistant and susceptible M.tb in macrophages and act in additive fashion with the antibiotics isoniazid and rifampicin. This exciting work uncovers targeting the intrinsic apoptosis pathway as a promising approach for TB host-directed therapy. Since safety and activity studies are underway in cancer clinics for MCL-1 and BCL-2 inhibitors, we expect that re-purposing them for TB treatment should translate more readily and rapidly to the clinic. Thus, the work supports further development of this host-directed therapy approach to augment current TB treatment.
Article
Full-text available
The recent epidemic caused by aerosolized SARS-CoV-2 virus illustrates the importance and vulnerability of the mucosal epithelial barrier against infection. Antimicrobial proteins and peptides (AMPs) are key to the epithelial barrier, providing immunity against microbes. In primitive life forms, AMPs protect the integument and the gut against pathogenic microbes. AMPs have also evolved in humans and other mammals to enhance newer, complex innate and adaptive immunity to favor the persistence of commensals over pathogenic microbes. The canonical AMPs are helictical peptides that form lethal pores in microbial membranes. In higher life forms, this type of AMP is exemplified by the defensin family of AMPs. In epithelial tissues, defensins, and calprotectin (complex of S100A8 and S100A9) have evolved to work cooperatively. The mechanisms of action differ. Unlike defensins, calprotectin sequesters essential trace metals from microbes, which inhibits growth. This review focuses on defensins and calprotectin as AMPs that appear to work cooperatively to fortify the epithelial barrier against infection. The antimicrobial spectrum is broad with overlap between the two AMPs. In mice, experimental models highlight the contribution of both AMPs to candidiasis as a fungal infection and periodontitis resulting from bacterial dysbiosis. These AMPs appear to contribute to innate immunity in humans, protecting the commensal microflora and restricting the emergence of pathobionts and pathogens. A striking example in human innate immunity is that elevated serum calprotectin protects against neonatal sepsis. Calprotectin is also remarkable because of functional differences when localized in epithelial and neutrophil cytoplasm or released into the extracellular environment. In the cytoplasm, calprotectin appears to protect against invasive pathogens. Extracellularly, calprotectin can engage pathogen-recognition receptors to activate innate immune and proinflammatory mechanisms. In inflamed epithelial and other tissue spaces, calprotectin, DNA, and histones are released from degranulated neutrophils to form insoluble antimicrobial barriers termed neutrophil extracellular traps. Hence, calprotectin and other AMPs use several strategies to provide microbial control and stimulate innate immunity.
Article
Background and purpose: L. monocytogenes remain a leading cause of foodborne infection. Listeriolysin O (LLO), an indispensable virulence determinant involved in diverse pathogenic mechanisms of L. monocytogenes infection, represents a promising therapeutic target. In this study, we sought to identify an effective inhibitor of LLO pore formation and its mechanism of action in the treatment of L. monocytogenes infection. Experimental approach: Haemolysis assays were carried out to screen an effective LLO inhibitor. The interaction between candidate and LLO was investigated using surface plasmon resonance and molecular docking. The effect of candidate on LLO-mediated cytotoxicity, barrier disruption and immune response were investigated. Finally, the in vivo effect of candidate on mice challenged with L. monocytogenes was examined. Key results: Amentoflavone, a natural flavone present in traditional Chinese herbs, effectively inhibited LLO pore formation by engaging the residues Lys93, Asp416, Tyr469 and Lys505 in LLO. Amentoflavone dose-dependently reduced L. monocytogenes-induced cell injury in an LLO-dependent manner. In the Caco-2 monolayer model, amentoflavone maintained the integrity of the epithelial barrier exposed to LLO. Amentoflavone inhibited the inflammatory response evoked by L. monocytogenes in an LLO-dependent manner, and inhibition was attributed to ability to block perforation-associated K+ efflux and Ca2+ influx. In the mouse infection model, amentoflavone treatment significantly reduced bacterial burden and pathological lesions in target organs, with a significant increase in survival rate. Conclusions and implications: Amentoflavone reduced the pathogenicity of L. monocytogenes by specifically inhibiting LLO pore formation, and this may represent a potential treatment for L. monocytogenes infection.
Preprint
Currently, there are no approved drugs for the treatment of flavivirus infection. Accordingly, we tested the inhibitory effects of the novel θ-defensin retrocyclin-101 (RC-101) against flavivirus infection, and investigated the mechanism underlying the potential inhibitory effects. First, RC-101 robustly inhibited both Japanese encephalitis virus (JEV) and Zika virus (ZIKV) infections. RC-101 exerted inhibitory effects on the entry and replication stages. Results also indicated that the non-structural protein NS2B-NS3 serine protease might serve as a potential viral target. Further, RC-101 inhibited protease activity at the micromolar level. We also demonstrated that with respect to the glycoprotein E protein of flavivirus, the DE loop of domain III, which is the receptor-binding domain of the E protein, might serve as another viral target of RC-101. Moreover, a JEV DE mutant exhibited resistance to RC-101, which was associated with deceased binding affinity of RC-101 to DIII. These findings provide a basis for the development of RC-101 as a potential candidate for the treatment of flavivirus infection. Importance RC has been reported to have a broad-spectrum antimicrobial activity. In this study, we firstly report that RC-101 could inhibit ZIKV and JEV infections. Moreover, both the NS2B-NS3 serine protease and the DE loop in the E glycoprotein might serve as the viral targets of RC-101.
Article
Full-text available
Innate and Adaptive Immune Responses during Listeria monocytogenes Infection, Page 1 of 2 Abstract It could be argued that we understand the immune response to infection with Listeria monocytogenes better than the immunity elicited by any other bacteria. L. monocytogenes are Gram-positive bacteria that are genetically tractable and easy to cultivate in vitro, and the mouse model of intravenous (i.v.) inoculation is highly reproducible. For these reasons, immunologists frequently use the mouse model of systemic listeriosis to dissect the mechanisms used by mammalian hosts to recognize and respond to infection. This article provides an overview of what we have learned over the past few decades and is divided into three sections: “Innate Immunity” describes how the host initially detects the presence of L. monocytogenes and characterizes the soluble and cellular responses that occur during the first few days postinfection; “Adaptive Immunity” discusses the exquisitely specific T cell response that mediates complete clearance of infection and immunological memory; “Use of Attenuated Listeria as a Vaccine Vector” highlights the ways that investigators have exploited our extensive knowledge of anti-Listeria immunity to develop cancer therapeutics.
Article
Objectives: To examine the effects and mechanisms of human neutrophil peptides in systemic infection and noninfectious inflammatory lung injury. Design: Prospective experimental study. Setting: University hospital-based research laboratory. Subjects: In vitro human cells and in vivo mouse models. Interventions: Wild-type (Friend virus B-type) and conditional leukocyte human neutrophil peptides transgenic mice were subjected to either sepsis induced by cecal ligation and puncture or acute lung injury by intratracheal instillation of hydrochloric acid followed by mechanical ventilation. Using human neutrophil peptides as bait, the basal cell adhesion molecule (CD239) and the purinergic P2Y purinoceptor 6 receptor were identified as the putative human neutrophil peptides receptor complex in human lung epithelial cells. Measurements and main results: In the cecal ligation and puncture sepsis model, Friend virus B-type mice exhibited higher systemic bacterial load, cytokine production, and lung injury than human neutrophil peptides transgenic mice. Conversely, an increased lung cytokine production was seen in Friend virus B-type mice, which was further enhanced in human neutrophil peptides transgenic mice in response to two-hit lung injury induced by hydrochloric acid and mechanical ventilation. The human neutrophil peptides-mediated inflammatory response was mediated through the basal cell adhesion molecule-P2Y purinoceptor 6 receptor signal pathway in human lung epithelial cells. Conclusions: Human neutrophil peptides are critical in host defense against infectious sepsis by their cationic antimicrobial properties but may exacerbate tissue injury when neutrophil-mediated inflammatory responses are excessive in noninfectious lung injury. Targeting the basal cell adhesion molecule/P2Y purinoceptor 6 signaling pathway may serve as a novel approach to attenuate the neutrophil-mediated inflammatory responses and injury while maintaining the antimicrobial function of human neutrophil peptides in critical illness.
Chapter
Listeria monocytogenes is an opportunistic intracellular bacterium responsible for the disease listeriosis. This review will update the knowledge on the four exotoxins secreted by this intracellular pathogen: the cholesterol-dependent cytolysin listeriolysin O (LLO), phosphatidylinositol-specific phospholipase C (PlcA), broad-range phospholipase C (PlcB), and hemolysin listeriolysin S (LLS). Each one of these exotoxins has evolved to perform specific and important functions in the extracellular or intracellular environment during the life cycle of L. monocytogenes. LLO, PlcA, and PlcB were discovered decades ago; however, recent studies are revisiting their functions and revealing new, previously unexpected insights. In the same line, LLS was discovered almost a decade ago, but it was recently deciphered that it is not only a toxin for eukaryotic cells but also a bacteriocin targeting bacteria closely related to L. monocytogenes. These latest findings, together with the knowledge generated during the history of listeriology, will be discussed in the light of their impact on the infectious process.
Article
Full-text available
Defensins as a prominent family of antimicrobial peptides (AMP) are major effectors of the innate immunity with a broad range of immune modulatory and antimicrobial activities. Particularly, defensins are the only recognized fast-response molecules that can neutralize a broad range of bacterial toxins, many of which are among the deadliest compounds on the planet. For a decade, the mystery of how a small and structurally conserved group of peptides can neutralize a heterogeneous group of toxins with little to no sequential and structural similarity remained unresolved. Recently, it was found that defensins recognize and target structural plasticity/thermodynamic instability, fundamental physicochemical properties that unite many bacterial toxins and distinguish them from the majority of host proteins. Binding of human defensins promotes local unfolding of the affected toxins, destabilizes their secondary and tertiary structures, increases susceptibility to proteolysis, and leads to their precipitation. While the details of toxin destabilization by defensins remain obscure, here we briefly review properties and activities of bacterial toxins known to be affected by or to be resilient to defensins and discuss how recognized features of defensins correlate with the observed inactivation.
Chapter
Listeria monocytogenes is an opportunistic intracellular bacterium responsible for the disease listeriosis. This review will update the knowledge on the four exotoxins secreted by this intracellular pathogen: the cholesterol-dependent cytolysin listeriolysin O (LLO), phosphatidylinositol-specific phospholipase C (PlcA), broad-range phospholipase C (PlcB), and hemolysin listeriolysin S (LLS). Each one of these exotoxins has evolved to perform specific and important functions in the extracellular or intracellular environment during the life cycle of L. monocytogenes. LLO, PlcA, and PlcB were discovered decades ago; however, recent studies are revisiting their functions and revealing new previously unexpected insights. In the same line, LLS was discovered almost a decade ago, but it was recently deciphered that it is not only a toxin for eukaryotic cells but also a bacteriocin targeting bacteria closely related to L. monocytogenes. These latest findings, together with the knowledge generated during the history of listeriology, will be discussed in the light of their impact on the infectious process.
Chapter
Listeria monocytogenes is an opportunistic intracellular bacterium responsible for the disease listeriosis. This review will update the knowledge on the four exotoxins secreted by this intracellular pathogen: the cholesterol-dependent cytolysin listeriolysin O (LLO), phosphatidylinositol-specific phospholipase C (PlcA), broad-range phospholipase C (PlcB), and hemolysin listeriolysin S (LLS). Each one of these exotoxins has evolved to perform specific and important functions in the extracellular or intracellular environment during the life cycle of L. monocytogenes. LLO, PlcA, and PlcB were discovered decades ago; however, recent studies are revisiting their functions and revealing new previously unexpected insights. In the same line, LLS was discovered almost a decade ago, but it was recently deciphered that it is not only a toxin for eukaryotic cells but also a bacteriocin targeting bacteria closely related to L. monocytogenes. These latest findings, together with the knowledge generated during the history of listeriology, will be discussed in the light of their impact on the infectious process.
Article
Phagocytes are crucial for host defense against bacterial pathogens. As first demonstrated by Metchnikoff, neutrophils and mononuclear phagocytes share the capacity to engulf, kill, and digest microbial invaders. Generally, neutrophils focus on extracellular, and mononuclear phagocytes on intracellular, pathogens. Reciprocally, extracellular pathogens often capitalize on hindering phagocytosis and killing of phagocytes, whereas intracellular bacteria frequently allow their engulfment and then block intracellular killing. As foreseen by Metchnikoff, phagocytes become highly versatile by acquiring diverse phenotypes, but still retaining some plasticity. Further, phagocytes engage in active crosstalk with parenchymal and immune cells to promote adjunctive reactions, including inflammation, tissue healing, and remodeling. This dynamic network allows the host to cope with different types of microbial invaders. Here we present an update of molecular and cellular mechanisms underlying phagocyte functions in antibacterial defense. We focus on four exemplary bacteria ranging from an opportunistic extracellular to a persistent intracellular pathogen.
Article
Antimicrobial peptides (AMPs) constitute an important part of innate immunity. AMP expression is often increased in response to colonic infection and inflammation. They also demonstrate a broad range of antimicrobial effects. Over the last two decades, the roles of antimicrobial peptides have been discovered and explored. Certain AMPs such as alpha defensin HD 5-6 and beta defensin HBD1 are constitutively expressed while others including defensin HBD2-4 and bactericidal/permeability increasing protein (BPI) are associated with Inflammatory Bowel Disease (IBD). Gene expression of several AMPs (beta defensin HBD2-4 and cathelicidin) is induced in response to invasion of gut microbes. Cathelicidin can directly modulate colitis while other AMPS such as lactoferrin and hepcidin are utilized as biomarkers of IBD disease activity. The application of AMPs for therapeutic purposes is still at an early stage of development. A few endogenous host-based AMPs (cathelicidin, elafin and SLPI) were shown to alter colitis when delivered intravenously or intracolonically in mouse colitis models. Novel AMPs (synthetic or artificial non-human peptides) with potent antimicrobial or anti-inflammatory property have been developed and may represent an alternative therapeutic approach against colitis and intestinal infection in the future. This report details the latest development of AMP-related research with emphasis in innate immunity and pathophysiology of colitis and intestinal infection.
Article
Full-text available
Antimicrobial peptides (AMPs) are evolutionarily ancient factors of the innate immune system that serve as a crucial first line of defense for humans, animals, and plants against infection. This review focuses on the structural organization, biosynthesis, and biological functions of AMPs that possess a β-hairpin spatial structure. Representatives of this class of AMPs are among the most active antibiotic molecules of animal origin. Due to their wide spectrum of activity and resistance to internal environmental factors, natural β-hairpin AMPbased compounds might become the most promising drug candidates.
Article
This was a 1060-g male stillborn at 27+ weeks of gestation born to a 35-year-old gravida 4, para 2 mother. The mother's pregnancy was uncomplicated until the onset of flulike symptoms and severe headache 8 days before delivery. She was treated symptomatically, and all her symptoms resolved within 2 days. Two days before delivery, she presented to her obstetrician's office complaining of decreased fetal movement. On admission, she was found to have a high fever and fetal demise. After induction, she was delivered of the stillborn. The placenta and the tissues of the stillborn had numerous microabscesses involving almost every site. Postmortem blood and various organ cultures grew Listeria monocytogenes. The mother's blood culture was also positive for the same organism. After being treated with antibiotics, she made a full recovery. The source and the nature of the food responsible for this case could not be identified. The purposes of this article were to review the characteristics of L. monocytogenes and the pathogenesis of the disease process and to familiarize the reader with the typical pathologic findings in the placenta and postmortem examination.
Article
Cyclische Peptide werden in vielen verschiedenen Organismen vorgefunden und zeichnen sich durch ihre Stabilität und ihre Rolle bei Abwehrmechanismen aus. Warum wurde in Säugetieren nur eine Klasse cyclischer Peptide gefunden? Möglicherweise haben wir noch nicht gründlich genug nach ihnen geforscht, oder die für ihre Erkennung notwendigen Techniken sind noch nicht vollständig entwickelt. Wir verstehen auch ihre faszinierende Biosynthese aus zwei separaten Genprodukten noch nicht. Die Bewältigung dieser Schwierigkeiten wird die Anwendung chemischer Hilfsmittel sowie Erkenntnisse über andere Klassen cyclischer Peptide erfordern. Wir stellen hier die neuesten Entwicklungen bei der Charakterisierung von Theta-Defensinen vor und beschreiben die wichtige Rolle, die die Chemie bei der Skizzierung ihrer Wirkmechanismen spielte. Darüber hinaus betonen wir das Potenzial von Theta-Defensinen als antimikrobielle Substanzen und als Gerüste für Peptidwirkstoffe.
Article
Antimicrobial peptides (AMPs) are important components of innate immunity. They are often expressed in response to colonic inflammation and infection. Over the last several years, the roles of several antimicrobial peptides have been explored. Gene expression of many AMPs (beta defensin HBD2-4 and cathelicidin) is induced in response to invasion of gut microbes into the mucosal barrier. Some AMPs are expressed in a constitutive manner (alpha defensin HD 5-6 and beta defensin HBD1), while others (defensin and bactericidal/permeability increasing protein BPI) are particularly associated with Inflammatory Bowel Disease (IBD) due to altered defensin expression or development of autoantibodies against Bactericidal/permeability increasing protein (BPI). Various AMPs have different spectrum and strength of antimicrobial effects. Some may play important roles in modulating the colitis (cathelicidin) while others (lactoferrin, hepcidin) may represent biomarkers of disease activity. The use of AMPs for therapeutic purposes is still at an early stage of development. A few natural AMPs were shown to be able to modulate colitis when delivered intravenously or intracolonically (cathelicidin, elafin and SLPI) in mouse colitis models. New AMPs (synthetic or artificial non-human peptides) are being developed and may represent new therapeutic approaches against colitis. This review discusses the latest research developments in the AMP field with emphasis in innate immunity and pathophysiology of colitis.
Article
Full-text available
Hyaluronan (HA) is a glycosaminoglycan polymer found in the extracellular matrix of virtually all mammalian tissues. Recent work has suggested a role for small, fragmented HA polymers in initiating innate defense responses in immune cells, endothelium, and epidermis through interaction with innate molecular pattern recognition receptors, such as TLR4. Despite these advances, little is known regarding the effect of fragmented HA at the intestinal epithelium, where numerous pattern recognition receptors act as sentinels of an innate defense response that maintains epithelial barrier integrity in the presence of abundant and diverse microbial challenges. Here we report that HA fragments promote expression of the innate antimicrobial peptide human β-defensin 2 (HβD2) in intestinal epithelial cells. Treatment of HT-29 colonic epithelial cells with HA fragment preparations resulted in time- and dose-dependent up-regulated expression of HβD2 protein in a fragment size-specific manner, with 35-kDa HA fragment preparations emerging as the most potent inducers of intracellular HβD2. Furthermore, oral administration of specific-sized HA fragments promotes the expression of an HβD2 ortholog in the colonic epithelium of both wild-type and CD44-deficient mice but not in TLR4-deficient mice. Together, our observations suggest that a highly size-specific, TLR4-dependent, innate defense response to fragmented HA contributes to intestinal epithelium barrier defense through the induction of intracellular HβD2 protein.
Article
Full-text available
Anthrax caused by Bacillus anthracis represents a major bioterroristic threat. B. anthracis produces lethal toxin (LeTx), a combination of lethal factor (LF) and protective antigen that plays a major role in anthrax pathogenesis. We demonstrate that human neutrophil α-defensins are potent inhibitors of LF. The inhibition of LF by human neutrophil protein (HNP-1) was noncompetitive. HNP-1 inhibited cleavage of a mitogen-activated protein kinase kinase and restored impaired mitogen-activated protein kinase signaling in LeTx-treated macrophages. HNP-1 rescued murine macrophages from B. anthracis-induced cytotoxicity, and in vivo treatment with HNP-1-3 protected mice against the fatal consequences of LeTx.
Article
Full-text available
Paneth cells at the base of small intestinal crypts secrete microbicidal α-defensins, termed cryptdins (Crps) in mice, as mediators of innate immunity. Proteomic studies show that five abundant Paneth cell α-defensins in C57BL/6 mice are strain specific in that they have not been identified in other inbred strains of mice. Two C57BL/6-specific peptides are coded for by the Defcr20 and -21 genes evident in the NIH C57BL/6 genome but absent from the Celera mixed-strain assembly, which excludes C57BL/6 data and differs from the NIH build with respect to the organization of the α-defensin gene locus. Conversely, C57BL/6 mice lack the Crp1, -2, -4, and -6 peptides and their corresponding Defcr1, -2, -4, and -6 genes, which are common to several mouse strains, including those of the Celera assembly. In C57BL/6 mice, α-defensin gene diversification appears to have occurred by tandem duplication of a multigene cassette that was not found in the mixed-strain assembly. Both mouse genome assemblies contain conserved α-defensin pseudogenes that are closely related to functional myeloid α-defensin genes in the rat, suggesting that the neutrophil α-defensin defect in mice resulted from progressive gene loss. Given the role of α-defensins in shaping the composition of the enteric microflora, such polymorphisms may influence outcomes in mouse models of disease or infection.
Article
Full-text available
Defensins are effectors of the innate immune response with potent antibacterial activity. Their role in antiviral immunity, particularly for non-enveloped viruses, is poorly understood. We recently found that human alpha-defensins inhibit human adenovirus (HAdV) by preventing virus uncoating and release of the endosomalytic protein VI during cell entry. Consequently, AdV remains trapped in the endosomal/lysosomal pathway rather than trafficking to the nucleus. To gain insight into the mechanism of defensin-mediated neutralization, we analyzed the specificity of the AdV-defensin interaction. Sensitivity to alpha-defensin neutralization is a common feature of HAdV species A, B1, B2, C, and E, whereas species D and F are resistant. Thousands of defensin molecules bind with low micromolar affinity to a sensitive serotype, but only a low level of binding is observed to resistant serotypes. Neutralization is dependent upon a correctly folded defensin molecule, suggesting that specific molecular interactions occur with the virion. CryoEM structural studies and protein sequence analysis led to a hypothesis that neutralization determinants are located in a region spanning the fiber and penton base proteins. This model was supported by infectivity studies using virus chimeras comprised of capsid proteins from sensitive and resistant serotypes. These findings suggest a mechanism in which defensin binding to critical sites on the AdV capsid prevents vertex removal and thereby blocks subsequent steps in uncoating that are required for release of protein VI and endosomalysis during infection. In addition to informing the mechanism of defensin-mediated neutralization of a non-enveloped virus, these studies provide insight into the mechanism of AdV uncoating and suggest new strategies to disrupt this process and inhibit infection.
Article
Full-text available
Defensins and LL-37 play key roles in maintaining mucosal barriers against invasive infection and initiating immune responses after infection or injury to mucosal surfaces. Defensins are widespread in nature and have activity against a broad range of pathogens. Defensins have direct antimicrobial effects and also modulate innate and adaptive immune responses. We consider the role of human defensins and the cathelicidin LL-37 in defense of respiratory, gastrointestinal, and genitourinary tracts and the oral cavity, skin, and eye. Human β-defensins (hBDs) and human defensins 5 and 6 (HD5 and −6) are involved most obviously in mucosal responses, as they are produced principally by epithelial cells. Human α-defensins 1–4 (or HNPs 1–4) are produced principally by neutrophils recruited to the mucosa. Understanding the biology of defensins and LL-37 is the beginning to clarify the pathophysiology of mucosal inflammatory and infectious diseases (e.g., Crohn’s disease, atopic dermatitis, lung or urinary infections). Challenges for these studies are the redundancy of innate defense mechanisms and the presence and interactions of many innate defense proteins in mucosal secretions.
Article
Full-text available
Many pathogenic gram-positive bacteria release exotoxins that belong to the family of cholesterol-dependent cytolysins. Here, we report that human alpha-defensins HNP-1 to HNP-3 acted in a concentration-dependent manner to protect human red blood cells from the lytic effects of three of these exotoxins: anthrolysin O (ALO), listeriolysin O, and pneumolysin. HD-5 was very effective against listeriolysin O but less effective against the other toxins. Human alpha-defensins HNP-4 and HD-6 and human beta-defensin-1, -2, and -3 lacked protective ability. HNP-1 required intact disulfide bonds to prevent toxin-mediated hemolysis. A fully linearized analog, in which all six cysteines were replaced by aminobutyric acid (Abu) residues, showed greatly reduced binding and protection. A partially unfolded HNP-1 analog, in which only cysteines 9 and 29 were replaced by Abu residues, showed intact ALO binding but was 10-fold less potent in preventing hemolysis. Surface plasmon resonance assays revealed that HNP-1 to HNP-3 bound all three toxins at multiple sites and also that solution-phase HNP molecules could bind immobilized HNP molecules. Defensin concentrations that inhibited hemolysis by ALO and listeriolysin did not prevent these toxins from binding either to red blood cells or to cholesterol. Others have shown that HNP-1 to HNP-3 inhibit lethal toxin of Bacillus anthracis, toxin B of Clostridium difficile, diphtheria toxin, and exotoxin A of Pseudomonas aeruginosa; however, this is the first time these defensins have been shown to inhibit pore-forming toxins. An "ABCDE mechanism" that can account for the ability of HNP-1 to HNP-3 to inhibit so many different exotoxins is proposed.
Article
Full-text available
In acute inflammation, infiltrating polymorphonuclear leukocytes (also known as PMNs) release preformed granule proteins having multitudinous effects on the surrounding environment. Here we present what we believe to be a novel role for PMN-derived proteins in bacterial phagocytosis by both human and murine macrophages. Exposure of macrophages to PMN secretion markedly enhanced phagocytosis of IgG-opsonized Staphylococcus aureus both in vitro and in murine models in vivo. PMN secretion activated macrophages, resulting in upregulation of the Fcgamma receptors CD32 and CD64, which then mediated the enhanced phagocytosis of IgG-opsonized bacteria. The phagocytosis-stimulating activity within the PMN secretion was found to be due to proteins released from PMN primary granules; thorough investigation revealed heparin-binding protein (HBP) and human neutrophil peptides 1-3 (HNP1-3) as the mediators of the macrophage response to PMN secretion. The use of blocking antibodies and knockout mice revealed that HBP acts via beta2 integrins, but the receptor for HNP1-3 remained unclear. Mechanistically, HBP and HNP1-3 triggered macrophage release of TNF-alpha and IFN-gamma, which acted in an autocrine loop to enhance expression of CD32 and CD64 and thereby enhance phagocytosis. Thus, we attribute what may be a novel role for PMN granule proteins in regulating the immune response to bacterial infections.
Article
Full-text available
We have been exploring the role of iron in the pathogenesis of the intracellular bacterial pathogen Legionella pneumophila. In previous studies, we have demonstrated that L. pneumophila intracellular multiplication in human monocytes is iron dependent and that IFN gamma-activated monocytes inhibit L. pneumophila intracellular multiplication by limiting the availability of iron. In this study, we have investigated the effect on L. pneumophila intracellular multiplication of lactoferrin, an iron-binding protein which is internalized via specific receptors on monocytes, and of nonphysiologic iron chelates which enter monocytes by a receptor-independent route. Apolactoferrin completely inhibited L. pneumophila multiplication in nonactivated monocytes, and enhanced the capacity of IFN gamma-activated monocytes to inhibit L. pneumophila intracellular multiplication. In contrast, iron-saturated lactoferrin had no effect on the already rapid rate of L. pneumophila multiplication in nonactivated monocytes. Moreover, it reversed the capacity of activated monocytes to inhibit L. pneumophila intracellular multiplication, demonstrating that L. pneumophila can utilize iron from the lactoferrin-lactoferrin receptor pathway. The capacity of iron-lactoferrin to reverse monocyte activation was dependent upon its percent iron saturation and not just its total iron content. Similarly, the nonphysiologic iron chelates ferric nitrilotriacetate and ferric ammonium citrate completely reverse and ferric pyrophosphate partially reversed the capacity of IFN gamma-activated monocytes to inhibit L. pneumophila intracellular multiplication, demonstrating that L. pneumophila can utilize iron derived from nonphysiologic iron chelates internalized by monocytes independently of the transferrin and lactoferrin endocytic pathways. This study suggests that at sites of inflammation, lactoferrin may inhibit or promote L. pneumophila intracellular multiplication in mononuclear phagocytes depending upon its degree of iron saturation. In addition, this study suggests a potential role for PMN in host defense against L. pneumophila--providing apolactoferrin to infected monocytes--and it supports the concept that PMN and monocytes may cooperate in host defense against intracellular parasites and other pathogens.
Article
Full-text available
Incubation of murine bone marrow-derived macrophages (BMM) in medium containing recombinant macrophage colony-stimulating factor (rM-CSF) stimulated influx, efflux, and the net accumulation of the fluid-phase pinocytic marker, lucifer yellow (LY). Stimulation was dose dependent, occurred within 5 min of addition of the growth factor, and was sustained. Previous experiments had shown that BMM treated with PMA were stimulated to accumulate LY, but compared with rM-CSF-treated cells, the onset of stimulation in PMA-treated macrophages was slower. In further comparisons of rM-CSF- and PMA-stimulated LY accumulation, it was found that rM-CSF-stimulated pinocytosis could be abolished by pretreatment with 0.5 mg/ml trypsin, whereas neither unstimulated nor PMA-stimulated LY accumulation was affected by trypsin pretreatment. These findings indicate that the rM-CSF response was initiated at the cell surface, while the PMA response occurred via intracellular (or trypsin-resistant) receptors. However, once initiated, the pinocytic responses elicited by either agent were very similar. First, rM-CSF-treated cells, like PMA-treated cells, showed extensive ruffling and formation of large phase-bright pinosomes. Second, both rM-CSF- and PMA-stimulated LY accumulation could be inhibited by treatment of cells with the cytoskeleton destabilizing drugs nocodazole, colchicine, or cytochalasin D. Finally, rM-CSF, like PMA, was found to stimulate efflux of LY from cells preloaded with the dye. Thus, both rM-CSF and PMA stimulate the net rate of solute flow through the macrophage endocytic compartment.
Article
Full-text available
Listeria monocytogenes insertion mutants defective in hemolysin production were generated using the conjugative transposons Tn916 and Tn1545. All of the nonhemolytic mutants (hly-) lacked a secreted 58-kD polypeptide, presumedly hemolysin, and were avirulent in a mouse model. An intracellular multiplication assay was established in monolayers of mouse bone marrow-derived macrophages, the J774 macrophage-like cell line, the CL.7 embryonic mouse fibroblast cell line, and the Henle 407 human epithelial cell line. The hly+ strain grew intracellularly in all of the tissue culture cells with a doubling time of approximately 60 min. In contrast, the hly- mutants failed to grow in the murine-derived tissue culture cells, but retained the ability to grow in the human tissue culture cells examined. Hemolytic-positive revertants were selected after passage of the hly- mutants through monolayers of J774 cells. In each case, the hemolytic revertants possessed the 58-kD polypeptide, were capable of intracellular growth in tissue culture monolayers and were virulent for mice.
Article
Full-text available
Recent studies have established the importance of neutrophils in innate resistance to Listeria monocytogenes infection in mice. The purpose of this study was to determine the importance of neutrophils in acquired resistance to L. monocytogenes infection. Previously immunized mice that were depleted of neutrophils by administration of the antigranulocyte monoclonal antibody RB6-8C5 demonstrated less resistance to L. monocytogenes challenge than did nonimmunized control mice. In contrast, immunized control mice exhibited a heightened resistance to rechallenge, as expected. These results suggest that neutrophils make previously unrecognized contributions to acquired immunity to a facultative intracellular pathogen.
Article
Full-text available
This study shows that in mice selectively depleted of neutrophils by treatment with a monoclonal antibody, RB6-8C5, listeriosis is severely exacerbated in the liver, but not in the spleen or peritoneal cavity during the crucial first day of infection. At sites of infection in the livers of neutrophil-depleted mice, Listeria monocytogenes grew to large numbers inside hepatocytes. By contrast, in the livers of normal mice neutrophils rapidly accumulated at infectious foci and this was associated with the lysis of infected hepatocytes that served to abort infection in these permissive cells. In the spleen the situation was different, in that depletion of neutrophils did not result in appreciable exacerbation of infection. In this organ intact infected cells, many of which appeared to be fibroblast-like stromal cells, were found at foci of infection in the presence or absence of large numbers of neutrophils. This suggests that neutrophils are less effective at destroying L. monocytogenes-infected target cells in the spleen than in the liver. Consequently, at least during the first day, the organism remained free to multiply intracellularly in the spleen in cells that are permissive for its growth. Presumably, the same situation exists in the peritoneal cavity, because depleting neutrophils did not severely exacerbate infection initiated at this site.
Article
Full-text available
The gram-positive bacterium Listeria monocytogenes is the causative agent of listeriosis, a highly fatal opportunistic foodborne infection. Pregnant women, neonates, the elderly, and debilitated or immunocompromised patients in general are predominantly affected, although the disease can also develop in normal individuals. Clinical manifestations of invasive listeriosis are usually severe and include abortion, sepsis, and meningoencephalitis. Listeriosis can also manifest as a febrile gastroenteritis syndrome. In addition to humans, L. monocytogenes affects many vertebrate species, including birds. Listeria ivanovii, a second pathogenic species of the genus, is specific for ruminants. Our current view of the pathophysiology of listeriosis derives largely from studies with the mouse infection model. Pathogenic listeriae enter the host primarily through the intestine. The liver is thought to be their first target organ after intestinal translocation. In the liver, listeriae actively multiply until the infection is controlled by a cell-mediated immune response. This initial, subclinical step of listeriosis is thought to be common due to the frequent presence of pathogenic L. monocytogenes in food. In normal individuals, the continual exposure to listerial antigens probably contributes to the maintenance of anti-Listeria memory T cells. However, in debilitated and immunocompromised patients, the unrestricted proliferation of listeriae in the liver may result in prolonged low-level bacteremia, leading to invasion of the preferred secondary target organs (the brain and the gravid uterus) and to overt clinical disease. L. monocytogenes and L. ivanovii are facultative intracellular parasites able to survive in macrophages and to invade a variety of normally nonphagocytic cells, such as epithelial cells, hepatocytes, and endothelial cells. In all these cell types, pathogenic listeriae go through an intracellular life cycle involving early escape from the phagocytic vacuole, rapid intracytoplasmic multiplication, bacterially induced actin-based motility, and direct spread to neighboring cells, in which they reinitiate the cycle. In this way, listeriae disseminate in host tissues sheltered from the humoral arm of the immune system. Over the last 15 years, a number of virulence factors involved in key steps of this intracellular life cycle have been identified. This review describes in detail the molecular determinants of Listeria virulence and their mechanism of action and summarizes the current knowledge on the pathophysiology of listeriosis and the cell biology and host cell responses to Listeria infection. This article provides an updated perspective of the development of our understanding of Listeria pathogenesis from the first molecular genetic analyses of virulence mechanisms reported in 1985 until the start of the genomic era of Listeria research.
Article
Full-text available
We purified two new minidefensins (RTD-2 and RTD-3) from the bone marrow of rhesus monkeys. Both were circular octadecapeptides that contained three intramolecular disulfide bonds and were homologous to RTD-1, a circular (theta) defensin previously described by Tang et al. (Science, 286, 498-502, 1999). However, whereas the 18 residues of RTD-1 represent spliced nonapeptide fragments derived from two different demidefensin precursors, RTD-2 and -3 comprise tandem nonapeptide repeats derived from only one of the RTD-1 precursors. Thus, circular minidefensins are products of a novel posttranslational system that generates effector molecule diversity without commensurate genome expansion. A system wherein two demidefensin genes can produce three circular minidefensins might allow n such genes to produce (n/2)(n+1) peptides.
Article
Full-text available
Human bone marrow expresses a pseudogene that encodes an antimicrobial peptide homologous to rhesus monkey circular minidefensins (delta-defensins). We prepared the putative ancestral human peptide by solid-phase synthesis and named it "retrocyclin." Retrocyclin did not cause direct inactivation of HIV-1, and its modest antibacterial properties resembled those of its rhesus homologs. Nevertheless, retrocyclin had a remarkable ability to inhibit proviral DNA formation and to protect immortalized and primary human CD4(+) lymphocytes from in vitro infection by both T-tropic and M-tropic strains of HIV-1. Confocal fluorescent microscopy studies performed with BODIPY-FL-labeled RC-101, a close analog of retrocyclin, showed that the peptide formed patch-like aggregates on the surface of CD4(+) cells. These findings suggest that retrocyclin interferes with an early stage of HIV-1 infection and that retrocyclin-like agents might be useful topical agents to prevent sexually acquired HIV-1 infections.
Article
Full-text available
Listeria monocytogenes is a facultative intracellular bacterial pathogen that escapes from a phagosome and grows in the host cell cytosol. The pore-forming cholesterol-dependent cytolysin, listeriolysin O (LLO), mediates bacterial escape from vesicles and is approximately 10-fold more active at an acidic than neutral pH. By swapping dissimilar residues from a pH-insensitive orthologue, perfringolysin O (PFO), we identified leucine 461 as unique to pathogenic Listeria and responsible for the acidic pH optimum of LLO. Conversion of leucine 461 to the threonine present in PFO increased the hemolytic activity of LLO almost 10-fold at a neutral pH. L. monocytogenes synthesizing LLO L461T, expressed from its endogenous site on the bacterial chromosome, resulted in a 100-fold virulence defect in the mouse listeriosis model. These bacteria escaped from acidic phagosomes and initially grew normally in cells and spread cell to cell, but prematurely permeabilized the host membrane and killed the cell. These data show that the acidic pH optimum of LLO results from an adaptive mutation that acts to limit cytolytic activity to acidic vesicles and prevent damage in the host cytosol, a strategy also used by host cells to compartmentalize lysosomal hydrolases.
Article
Listeria monocytogenes insertion mutants defective in hemolysin production were generated using the conjugative transposons Tn916 and Tn1545. All of the nonhemolytic mutants (hly-) lacked a secreted 58-kD polypeptide, presumedly hemolysin, and were avirulent in a mouse model. An intracellular multiplication assay was established in monolayers of mouse bone marrow-derived macrophages, the J774 macrophage-like cell line, the CL.7 embryonic mouse fibroblast cell line, and the Henle 407 human epithelial cell line. The hly+ strain grew intracellularly in all of the tissue culture cells with a doubling time of approximately 60 min. In contrast, the hly- mutants failed to grow in the murine-derived tissue culture cells, but retained the ability to grow in the human tissue culture cells examined. Hemolytic-positive revertants were selected after passage of the hly- mutants through monolayers of J774 cells. In each case, the hemolytic revertants possessed the 58-kD polypeptide, were capable of intracellular growth in tissue culture monolayers and were virulent for mice.
Article
Murine listeriosis was introduced 35 years ago as a model with which to analyze mechanisms of antibacterial defense that are independent of antibodies. Listeria monocytogenes was shown to be an intramacrophage pathogen with capacity to induce the generation of a state of specific immunity in the form of DTH and a macrophage system with enhanced non-specific bactericidal activity. The demonstration that anti-Listeria immunity and DTH can be passively transferred with T cells was taken to indicate that the T cells responsible for DTH function upregulate the listericidal function of macrophages. This interpretation is contradicted by recent research showing that CD8 T cells, rather than CD4 T cells, are responsible for mediating adoptive immunity. However, T-cell depletion studies show that primary infection can eventually be resolved in the absence of either CD8 or CD4 T cells. On the other hand, infection becomes lethal in the absence of neutrophils or NK cells. It is apparent, therefore, that the most important defense against primary listeriosis resides with the functions of neutrophils and NK cells that are mobilized early in infection. Antigen-specific T cells function at a later time to resolve infection more efficiently. It is apparent that T cells are much more important in defense against secondary infection.
Article
The first cyclic peptide discovered in animals is an antimicrobial octadecapeptide that is expressed in leukocytes of rhesus monkeys. The peptide, termed rhesus theta-defensin 1 (RTD-1) is the prototype of a new family of antimicrobial peptides, which like the previously characterized alpha- and beta-defensin families, possesses broad spectrum microbicidal activities against bacteria, fungi, and protects mononuclear cells from infection by HIV-1. The cyclic theta-defensin structure is essential for a number of its antimicrobial properties, as demonstrated by the markedly reduced microbicidal activities of de-cyclized theta-defensin analogs. Genetic and biochemical experiments disclosed that the biosynthesis of RTD-1 results from the head-to-tail joining of two nine-amino acid peptides, each of which is donated by a separate precursor polypeptide, which are in fact C-terminally truncated pro-alpha-defensins. Alternate combinations of the two nonapeptides generate two additional macaque theta-defensins, RTD-2 and RTD-3. Humans do not express theta-defensin peptides, but mRNAs encoding at least two theta-defensins are expressed in human bone marrow. However, in each case the open reading frame is interrupted by a stop codon in the signal peptide-coding region. The mature theta-defensin peptide is a two-stranded beta-sheet that, like the alpha- and beta-defensins, is stabilized by three disulfides. However, the parallel orientation of the theta-defensin disulfide arrangement allows for substantial flexibility around its short axis. Unlike alpha- and beta-defensins, RTD-1 lacks an amphiphilic topology. This may partially explain the unusual interaction between theta-defensins and phospholipid bilayers.
Article
A procedure has been developed for electroporation-mediated transformation of Listeria monocytogenes with plasmid DNA. The method was optimized for intact cells of L. monocytogenes 23074 by determining the effects of field strength, cell density, and plasmid DNA topology. Transformation efficiencies were dramatically increased when cells were treated with penicillin. Optimum frequencies of transformation (4 × 106 transformation/μg DNA) were obtained when cells were grown in 10 μg/ml of penicillin G and electroporated at a field strength of 10kV/cm. Using, this procedure, transformation of relaxedf plasmid DNA from ligation reactions provided 1 × 104 transformants/μg DNA, allowing direct molecular cloning of DNA into this organism.
Article
This review rather than covering the whole field intends to highlight recent findings on the Listeria monocytogenes infectious process or some Listeria specific traits, place them within the framework of well-established data, and demonstrate how this Gram-positive bacterium has, in two decades, emerged as a multifaceted paradigm. Indeed, the cell biology of the infectious process has been deciphered in great detail and provided insights in both the way bacterial pathogen manipulate the host and unsuspected functions of well-known cellular proteins. The intra- and intercellular motility has in particular been instrumental in understanding actin-based motility in general. The analysis of the two main Listeria invasion proteins and that of their host specificities have illustrated how in vitro studies can help generating or choosing relevant animal models for in vivo studies. Listeria post-genomics studies have highlighted the power of comparative genomics in virulence studies. Together, Listeria, after being recognized as a powerful tool in immunology, now appears as one of the most insightful models in infection biology.
Article
The antimicrobial effector activity of phagocytes is crucial in the host innate defense against infection, and the classic view is that the phagocytes operating against intracellular and extracellular microbial pathogens are,respectively, macrophages and neutrophils. As a result of the common origin of the two phagocytes, they share several functionalities, including avid phagocytosis,similar kinetic behavior under inflammatory/infectious conditions, and antimicrobial and immunomodulatory activities. However, consequent to specialization during their differentiation, macrophages and neutrophils acquire distinctive, complementary features that originate different levels of antimicrobial capacities and cytotoxicity and different tissue localization and lifespan.This review highlights data suggesting the perspective that the combination of overlapping and complementary characteristics of the two professional phagocytes promotes their cooperative participation as effectors and modulators in innate immunity against infection and as orchestrators of adaptive immunity. In the concerted activities operating in antimicrobial innate immunity, macrophages and neutrophils are not able to replace each other. The common and complementary developmental,kinetic, and functional properties of neutrophils and macrophages make them the effector arms of a myeloid phagocyte system that groups neutrophils with members of the old mononuclear phagocyte system. The use by mammals of a system with two dedicated phagocytic cells working cooperatively represents an advantageous innate immune attack strategy that allows the efficient and safe use of powerful but dangerous microbicidal molecules.This crucial strategy is a target of key virulence mechanisms of successful pathogens.
Article
We describe an automated fluorescence microscopy-based assay that quantifies the invasion of mammalian cells by intracellular pathogens. Pathogens associated with host cell surfaces, intracellular pathogens and mammalian cells are directly counted based on their specific fluorescent labeling. Such approach utilizes automated image acquisition and processing, and is thus ideally suited for high-throughput analyses. This method was validated using Listeria monocytogenes as a model intracellular pathogen.
Article
Whereas significant knowledge is accumulating on the antibacterial and antifungal properties of host defense peptides (HDPs) and their synthetic mimics, much less is known of their activities against parasites. A variety of in vitro and in vivo antiparasitic assays suggest that these notorious antimicrobial compounds could represent a powerful tool for the development of novel drugs to fight parasites in the vertebrate host or to complement current therapeutic strategies, albeit the fact that HDPs essentially act by nonspecific mechanisms casts serious doubt on their ability to exert sufficient selectivity to be considered ideal candidates for drug development. This minireview summarizes recent efforts to assess the antiparasitic properties of HDPs and their synthetic derivatives, focusing on two of the most used models - Plasmodium and Leishmania species - for antiparasitic assays against the different development stages.
Article
There is widespread acceptance that cationic antimicrobial peptides, apart from their membrane-permeabilizing/disrupting properties, also operate through interactions with intracellular targets, or disruption of key cellular processes. Examples of intracellular activity include inhibition of DNA and protein synthesis, inhibition of chaperone-assisted protein folding and enzymatic activity, and inhibition of cytoplasmic membrane septum formation and cell wall synthesis. The purpose of this minireview is to question some widely held views about intracellular-targeting antimicrobial peptides. In particular, I focus on the relative contributions of intracellular targeting and membrane disruption to the overall killing strategy of antimicrobial peptides, as well as on mechanisms whereby some peptides are able to translocate spontaneously across the plasma membrane. Currently, there are no more than three peptides that have been convincingly demonstrated to enter microbial cells without the involvement of stereospecific interactions with a receptor/docking molecule and, once in the cell, to interfere with cellular functions. From the limited data currently available, it seems unlikely that this property, which is isolated in particular peptide families, is also shared by the hundreds of naturally occurring antimicrobial peptides that differ in length, amino acid composition, sequence, hydrophobicity, amphipathicity, and membrane-bound conformation. Microbial cell entry and/or membrane damage associated with membrane phase/transient pore or long-lived transitions could be a feature common to intracellular-targeting antimicrobial peptides and mammalian cell-penetrating peptides that have an overrepresentation of one or two amino acids, i.e. Trp and Pro, His, or Arg. Differences in membrane lipid composition, as well as differential lipid recruitment by peptides, may provide a basis for microbial cell killing on one hand, and mammalian cell passage on the other.
Article
This study demonstrated that atypical virulent filaments of Listeria monocytogenes (rough variant type II and designated FR for this study), isolated from clinical specimens or generated during exposure to pulsed-plasma gas discharge in liquids, were shown to be capable of survival when engulfed by human polymorphonuclear leukocytes (PMNLs). Factors shown to significantly influence the maximal respiratory burst response in PMNLs and survival of different internalized cell or filament forms of L. monocytogenes were bacterial strain, culture form, degree of opsonization (with and without the use of 10% serum) and composition of the bacterial growth media used before uptake by PMNLs. Opsonized regular-sized L. monocytogenes cells grown on blood agar (BA) elicited the greatest respiratory burst response and survived best in PMNLs. The filamentous (FR) and multiple cell chain (MCR) rough variants were significantly less susceptible to uptake and survival in PMNLs. Supplementation of tryptone soya agar with hemin resulted in significantly reduced chemiluminescence responses in phagocytosing PMNLs compared with the maximal levels observed from prior bacterial growth on BA or brain heart infusion agar that also contained a source of iron. The MCR variants secreting decreased levels of a peptidoglycan hydrolase CwhA protein exhibited the lowest percentage survival when internalized in PMNLs compared with wild-type smooth or FR culture variants as determined by the macrophage-killing assay.
Article
Infection is a major cause of death in patients with aplastic anemia (AA). There are differences between the immunocompromised state of a patient with AA and the patient who is neutropenic due to chemotherapy and this leads to a difference in the infections that they incur. Prolonged neutropenia is one of the largest risk factors for the development of infections with the invasive mycoses and bacteria. Recovery from neutropenia is directly related to survival, and supportive care plays a large role in protection while the patient is in a neutropenic state. The most common invasive mycoses include the Aspergillus species, Zygomycetes, Candida spp., and Fusarium spp. Bacterial infections that are seen in patients with AA include gram-positive coagulase-negative Staphylococcus species, Enterococcus, Staphylococus aureus, Clostridium spp., Micrococcus, alpha-hemolytic streptococci, Listeria monocytogenes, and Bacillus cereus. Gram-negative infections including gram-negative bacilli, Escherichia coli, Salmonella, Bacteroides fragilis, Klebsiella oxytoca, Klebsiella pneumonia, Aeromonas hydrophilia, Pseudomonas aeruginosa, and Vibrio vulnificus. Viral infections are much less common but include those that belong to the Herpesviridae family, community-acquired respiratory viral infection, and the viral hepatitides A, B, and C. Evidence of the parasite Strongyloides stercoralis has also been documented. This review discusses the major invasive fungal infections, bacterial pathogens, parasites, and viral infections that are found in patients with AA who are treated with immunosuppressive therapy. The specific immune impairment and current treatment parameters for each of these classes of infection will also be discussed.
Article
IFN-gamma plays a critical role during the immune response to infection with Listeria monocytogenes. Early in the innate response NK cells are thought to be a primary source of IFN-gamma; however, protection can be mediated by the presence of significant numbers of primed IFN-gamma-secreting CD8(+) T cells. In this report, we examined the early response to Listeria and found that 18 h after infection spleens contain CD11b(+), Gr-1(high), or Ly6G(+) cells that produce significant IFN-gamma. Morphological analysis of sorted Gr-1(high)IFN-gamma(+) and Gr-1(low)IFN-gamma(+) or Ly6G(+)IFN-gamma(+) cells confirmed that these cells were neutrophils. The importance of IFN-gamma production by these cells was further tested using adoptive transfer studies. Transfer of purified neutrophils from Ifng(+/+) mice led to increased bacterial clearance in Ifng(-/-) mice. Transfer of Ifng(-/-) neutrophils provided no such protection. We conclude that neutrophils are an early source of IFN-gamma during Listeria infection and are important in providing immune protection.
Article
We tested the ability of human peripheral blood monocytes to kill Candida albicans and Candida parapsilosis. Evidence that multiple fungicidal mechanisms operate in normla monocytes was found. Normal monocytes ingested and killed viable C. albicans, and could iodinate heat-killed C. albicans. Both functions were defective in monocytes from subjects with myeloperoxidase deficiency or chronic granulomatous disease. Methimazole, isoniazid, and aminotriazole inhibited iodination by normal monocytes without impairing their ability to kill C. albicans, indicating that iodination was not essential to the myeloperoxidase-hydrogen peroxide-mediated fungicidal system of the monocyte. C. parapsilosis, an organism killed with supranormal efficacy by monocytes from a patient with hereditary myeloperoxidase deficiency, was selected to examine the myeloperoxidase-independent fungicidal mechanisms of monocytes. Monocytes were obtained from the blood of normal or leukemic subjects and homogenized in 0.34 M sucrose to yield fractions rich in cytoplasmic granules. These fractions were extracted with 0.01 M citric acid and the soluble components were separated by micropreparative polyacrylamide electrophoresis. Monocytes were found to contain cationic proteins, other than myeloperoxidase, that kill C. parapsilosis in vitro.
Article
Defensins are broad-spectrum antimicrobial peptides that are abundant in human, rat, and rabbit neutrophils. We now report that neutrophils from nine strains of mice lacked appreciable defensin content. Mice may therefore be imperfect experimental surrogates for humans or rats in models of infection in which neutrophil function is significant.
Article
Defensins (molecular weight 3500 to 4000) act in the mammalian immune response by permeabilizing the plasma membranes of a broad spectrum of target organisms, including bacteria, fungi, and enveloped viruses. The high-resolution crystal structure of defensin HNP-3 (1.9 angstrom resolution, R factor 0.19) reveals a dimeric beta sheet that has an architecture very different from other lytic peptides. The dimeric assembly suggests mechanisms by which defensins might bind to and permeabilize the lipid bilayer.
Article
Listeria monocytogenes was used as a model intracellular parasite to study stages in the entry, growth, movement, and spread of bacteria in a macrophage cell line. The first step in infection is phagocytosis of the Listeria, followed by the dissolution of the membrane surrounding the phagosome presumably mediated by hemolysin secreted by Listeria as nonhemolytic mutants remain in intact vacuoles. Within 2 h after infection, each now cytoplasmic Listeria becomes encapsulated by actin filaments, identified as such by decoration of the actin filaments with subfragment 1 of myosin. These filaments are very short. The Listeria grow and divide and the actin filaments rearrange to form a long tail (often 5 microns in length) extending from only one end of the bacterium, a "comet's tail," in which the actin filaments appear randomly oriented. The Listeria "comet" moves to the cell surface with its tail oriented towards the cell center and becomes incorporated into a cell extension with the Listeria at the tip of the process and its tail trailing into the cytoplasm behind it. This extension contacts a neighboring macrophage that phagocytoses the extension of the first macrophage. Thus, within the cytoplasm of the second macrophage is a Listeria with its actin tail surrounded by a membrane that in turn is surrounded by the phagosome membrane of the new host. Both these membranes are then solubilized by the Listeria and the cycle is repeated. Thus, once inside a host cell, the infecting Listeria and their progeny can spread from cell to cell by remaining intracellular and thus bypass the humoral immune system of the organism. To establish if actin filaments are essential for the spread of Listeria from cell to cell, we treated infected macrophages with cytochalasin D. The Listeria not only failed to spread, but most were found deep within the cytoplasm, rather than near the periphery of the cell. Thin sections revealed that the net of actin filaments is not formed nor is a "comet" tail produced.
Article
Functional myeloperoxidase contained in granules of polymorphonuclear neutrophil leukocytes or in fixed whole cells can be endocytosed by mouse peritoneal macrophages. Acquired myeloperoxidase was distributed in what we considered to be the secondary lysosomal system and, following a phagocytic stimulation, was delivered to newly formed phagosomes containing the targets.
Article
CD-1 mice inoculated intraperitoneally with Mycobacterium avium, M. bovis, M. microti or M. kansasii showed a persistent peritoneal granulocytosis (above 10(6) cells, i.e. more than 15% of total cells) throughout the 3 month period of infection studied. By contrast, in mice inoculated with the non-pathogenic M. aurum or with heat-killed M. avium the number of granulocytes decreased progressively after the first 15 days. No mycobacteria were found in granulocytes except in the first 2 days of infection. The mycobacteria-induced chronic granulocytosis was accompanied by phagocytosis of granulocytes by macrophages. Throughout the 3 months of infection, macrophages were found to contain intracellular lactoferrin. Macrophages with lactoferrin were also found in subcutaneous infection caused by M. marinum and in systemic infection caused by M. avium or M. kansasii. The in vitro activity of mouse peritoneal macrophages against M. avium and M. microti was increased after ingestion of granulocyte material by macrophages. These results lead us to propose that granulocytes participate in the host response to mycobacterial infections, not as phagocytes but rather through an indirect mechanism, as a source for the macrophages of molecules involved in antimicrobial mechanisms (e.g., lactoferrin and myeloperoxidase) lacking in the mature macrophage.
Article
Human polymorphonuclear leukocytes (PMN) contain three antimicrobial and cytotoxic peptides which belong to a family of mammalian granulocyte peptides named defensins. To determine their potential availability for extracellular microbicidal or cytotoxic events, we quantified the extracellular release of defensins after stimulation of human PMN with phorbol myristate acetate and opsonized zymosan. As determined by enzyme immunoassay and confirmed by polyacrylamide gel electrophoresis and densitometry, 10(6) human PMN contained 4 to 5 micrograms of defensins. After stimulation with a high concentration of phorbol myristate acetate (1 microgram/ml), about 8% of PMN defensins were found in the media. Release of defensins correlated best with the release of azurophil granule marker beta-glucuronidase or elastase and poorly with the release of either the specific granule marker lactoferrin or cytoplasmic lactate dehydrogenase. Phagocytosis of opsonized zymosan resulted in the extracellular release of less than 3% of PMN defensins. The factors responsible for less release of defensins into media relative to the release of other azurophil granule proteins may include heterogeneity of azurophil granules and the affinity of defensins for cellular surfaces and opsonized particles. In vivo, defensins are most likely to reach effective microbicidal or cytotoxic concentrations in PMN-rich exudates (pus), in confined environments of the phagolysosomes, or in intercellular clefts between PMN and their targets.
Article
Escherichia coli-induced acidic pH conditions were observed during the in vitro adherence of E. coli to HeLa cells. No pH changes occurred in the absence of adherence. This suggests that adherence affects the function or interaction of HeLa cells and E. coli.
Article
Listeriolysin O (LLO) is a pore-forming cytolysin that enables Listeria monocytogenes to escape from a host cell vacuole. The structural gene for the related cytolysin perfringolysin O (pfo) was cloned downstream from the promoter for hly, the gene encoding LLO, both on a plasmid and on the L. monocytogenes chromosome. Both strains secreted active PFO, although regulation was not identical to that of LLO. The chromosomal PFO-expressing strain was characterized for intracellular growth and cell-to-cell spread. It escaped from a host cell vacuole with 64% efficiency compared with the wild type as determined by immunofluorescent staining of bacteria for F-actin, a marker for entry into the cytoplasm. In addition, it replicated intracellularly with a doubling time similar to that of the wild type for 5 h, after which growth was aborted because of a cytotoxic effect on the host cell and influx of extracellular gentamicin. The chromosomal PFO strain was able to plaque in mouse L2 fibroblasts, but it did so at 20% efficiency compared with the wild type and the plaques were significantly smaller. Both strains expressing PFO were completely avirulent in mice. These results indicate that PFO can mediate escape from a host cell vacuole but cannot complement an hly deletion strain for virulence.
Article
The importance of neutrophils in killing extracellular, pyogenic bacteria has long been established. However, there is only indirect evidence for a role for neutrophils in resistance against intracellular organisms. In this study, we directly demonstrate the involvement of neutrophils in defense against Listeria monocytogenes in normal C.B-17 immunocompetent and C.B-17 SCID mice. Because of the lack of sterilizing T-cell immunity, SCID mice are unable to completely eliminate listeriae systemically and become chronically infected. Both immunocompetent and SCID mice treated with a specific neutrophil-depleting monoclonal antibody during the early stages of Listeria infection were rendered remarkably sensitive to the organism, with a high level of mortality resulting from enhanced bacterial growth. At a late stage of infection in SCID mice, however, administration of neutrophil-depleting antibody did not affect mortality. In spite of the neutrophil depletion, other parameters of nonspecific immune function were normal. Macrophage infiltration to the site of infection and macrophage expression of major histocompatibility complex class II molecules were unaffected. Moreover, NK cell functions were normal as measured by infiltration to an infection site and gamma interferon production. These data demonstrate an important role for neutrophils in controlling the acute phase of Listeria infection, cooperating with, and yet independent of, macrophages and NK cells.
Article
Porcine leukocytes contained three homologous peptides, PG-1, 2 and 3, that manifested potent microbicidal activity against Escherichia coli, Listeria monocytogenes and Candida albicans in vitro. The peptides ('protegrins') were composed of 16 (PG-2) or 18 amino acid residues, and, like tachyplesins (broad-spectrum antibiotic peptides of horseshoe crab hemocytes), they contained two intramolecular cystine disulfide bonds. Considerably smaller than defensins, protegrins nevertheless showed substantial homology to them, especially to the 'corticostatic' rabbit defensin, NP-3a. The relatively simple structure of protegrins should provide useful prototypes for constructing congeners with selectively enhanced host defense activities.
Article
We measured concentrations of defensins (human neutrophil peptides) in the plasma of healthy volunteers and patients with sepsis and meningitis. When a sensitive enzyme immunoassay was used, defensins were detected in plasma samples from 13 of 24 healthy blood donors, with a mean +/- SD of 42 +/- 53 ng/ml. Defensin levels in plasma samples from seven patients with sepsis at the onset of disease ranged from 900 ng/ml to 170,000 ng/ml. In 10 patients with meningitis in the initial phase of disease, plasma defensin concentrations ranged from 120 ng/ml to 910 ng/ml. Defensin concentrations in the plasma of both patient groups were significantly higher than those in healthy blood donors (p < 0.01), and patients with sepsis had higher defensin levels than patients with meningitis (p < 0.01). Defensin levels were significantly (p < 0.01) lower after the beginning of specific antibiotic therapy. Defensin concentrations in the plasma of patients with sepsis and meningitis correlated only weakly (r = 0.38) with blood neutrophil count. In vitro studies of defensin added to plasma indicated that all defensin was bound to plasma proteins. At high concentrations (1000 micrograms/ml), defensins caused precipitation of plasma proteins. Because plasma defensin levels may reflect neutrophil activation at sites of infection and inflammation, studies of the clinical utility of defensin ELISA are indicated.
Article
Defensins are antimicrobial and cytotoxic peptides that contain 29-35 amino acid residues, including six invariant cysteines whose intramolecular disulfide bonds cyclize and stabilize them in a complexly folded, triple-stranded beta-sheet configuration. Generated by the proteolytic processing of 93-95 amino acid precursor peptides, the constitute > 5% of the total cellular protein in human and rabbit neutrophils (polymorphonucleated neutrophils--PMN) and are also produced by rabbit lung macrophages and by mouse and rabbit small intestinal Paneth cells. Despite their prominence in rat PMN, defensins are not found in murine PMN. The antimicrobial spectrum of defensins includes gram positive and gram negative bacteria, mycobacteria, T. pallidum, many fungi, and some enveloped viruses. Defensins exert nonspecific cytotoxic activity against a wide range of normal and malignant targets, including cells resistant to TNF-alpha and NK-cytolytic factor. They appear to kill mammalian target cells and microorganisms by a common mechanism, which involves initial electrostatic interactions with negatively charged target cell surface molecules (likely the head groups of polar membrane lipids), followed by insertion into the cell membranes which they permeabilize, forming voltage-regulated channels. In addition to their antimicrobial and cytotoxic properties, some defensins act as opsonins, while others inhibit protein kinase C, bind specifically to the ACTH receptor and block steroidogenesis or act as selective chemoattractants for monocytes. Defensins are a newly delineated family of effector molecules whose contribution to host defense, inflammation, and cytotoxicity may be considerable for humans, even though it is unlikely to be revealed by experimentation with mice.
Article
Reports in the past few years have shown the involvement of different cells and cytokines in controlling the infection with the intracellular facultative pathogen Listeria monocytogenes. A synergistic interaction of T-cell-independent and -dependent processes takes place but the nature of these interactions and of the relevant cells and cytokines depends on both the stage of the infection and the tissue that is involved.
Article
Human neutrophils contain two structurally distinct types of antimicrobial peptides, beta-sheet defensins (HNP-1 to HNP-4) and the alpha-helical peptide LL-37. We used radial diffusion assays and an improved National Committee for Clinical Laboratory Standards-type broth microdilution assay to compare the antimicrobial properties of LL-37, HNP-1, and protegrin (PG-1). Although generally less potent than PG-1, LL-37 showed considerable activity (MIC, <10 microgram/ml) against Pseudomonas aeruginosa, Salmonella typhimurium, Escherichia coli, Listeria monocytogenes, Staphylococcus epidermidis, Staphylococcus aureus, and vancomycin-resistant enterococci, even in media that contained 100 mM NaCl. Certain organisms (methicillin-resistant S. aureus, Proteus mirabilis, and Candida albicans) were resistant to LL-37 in media that contained 100 mM NaCl but were susceptible in low-salt media. Burkholderia cepacia was resistant to LL-37, PG-1, and HNP-1 in low- or high-salt media. LL-37 caused outer and inner membrane permeabilization of E. coli ML-35p. Chromogenic Limulus assays revealed that LL-37 bound to E. coli O111:B4 lipopolysaccharide (LPS) with a high affinity and that this binding showed positive cooperativity (Hill coefficient = 2.02). Circular dichroism spectrometry disclosed that LL-37 underwent conformational change in the presence of lipid A, transitioning from a random coil to an alpha-helical structure. The broad-spectrum antimicrobial properties of LL-37, its presence in neutrophils, and its inducibility in keratinocytes all suggest that this peptide and its precursor (hCAP-18) may protect skin and other tissues from bacterial intrusions and LPS-induced toxicity. The potent activity of LL-37 against P. aeruginosa, including mucoid and antibiotic-resistant strains, suggests that it or related molecules might have utility as topical bronchopulmonary microbicides in cystic fibrosis.
Article
Analysis of rhesus macaque leukocytes disclosed the presence of an 18-residue macrocyclic, tridisulfide antibiotic peptide in granules of neutrophils and monocytes. The peptide, termed rhesus theta defensin-1 (RTD-1), is microbicidal for bacteria and fungi at low micromolar concentrations. Antibacterial activity of the cyclic peptide was threefold greater than that of an open-chain analog, and the cyclic conformation was required for antimicrobial activity in the presence of 150 millimolar sodium chloride. Biosynthesis of RTD-1 involves the head-to-tail ligation of two alpha-defensin-related nonapeptides, requiring the formation of two new peptide bonds. Thus, host defense cells possess mechanisms for synthesis and granular packaging of macrocyclic antibiotic peptides that are components of the phagocyte antimicrobial armamentarium.
Article
"... It is likely that the leukocyte granulations are in fact secretory products, which the cell dissolves and spreads to the environment as needed", Paul Ehrlich, 1900. Neutrophil granules have long been recognized as mediators of innate host defense. Newly discovered functions for individual granule proteins suggest that granule constituents may also participate in adaptive immune responses. Neutrophil granule-derived cathepsin G, azurocidin/CAP37 and alpha-defensins have been shown to be chemotactic for mononuclear cells and neutrophils. Analysis of the chemotactic activity of alpha-defensins shows that they induce CD45RA+ and CD8 T-lymphocyte cell migration at concentrations 10 to 100-fold below that required for direct bactericidal activity. Additionally, alpha and beta defensins form chemotactic gradients for immature dendritic cells. Recruiting immature dendritic cells to sites of infection is one way for neutrophil granule proteins to initiate adaptive immune responses. Granules found in other leukocytes such as mast cells also contain serine proteases, such as chymase, that are known to chemoattract neutrophils and mononuclear cells. Preliminary evidence suggests that exocytosis of granule-derived products from a variety of leukocytes can mobilize inflammatory cells and immunocytes. Thus, leukocyte granule-derived proteins, more rapidly than chemokines, can mobilize cells that mediate innate host defense and adaptive immunity.
Article
The cytoplasmic granules of mammalian neutrophils contain several antimicrobial peptides. Some, like defensins, are fully processed before storage, whereas others are stored as precursors that require additional processing. Cathelicidins are bipartite molecules with an N-terminal cathelin domain and an antimicrobial C-terminal domain. Humans apparently have only one cathelicidin gene. Its product, hCAP-18, is present in the secondary (specific) granules of neutrophils, and its C-terminal antimicrobial peptide, LL-37, is liberated by proteinase 3 coincident with degranulation and secretion. Many nonmyeloid tissues also express hCAP-18, including epididymis, spermatids, keratinocytes, epithelial cells, and various lymphocytes. LL-37 stimulates chemotaxis, acting via the formyl peptide-like receptor-1. The structurally diverse cathelicidin-derived antimicrobial peptides of animals provide interesting models for pharmaceutical development. PR-39, a proline-rich porcine cathelicidin, has shown efficacy in limiting myocardial damage after experimental ischemia in rodent models. Porcine protegrins are in stage III clinical trials to prevent oral mucositis caused by radiation or chemo-therapy.
Article
During the past year, novel beta-defensins of mice and men have been identified, together with a novel defensin subfamily (the circular or 'theta' minidefensins) in primates. Insight into the evolution of defensins has been obtained from structural studies, and several mechanisms related to microbial resistance to defensins have been delineated. There is now convincing evidence that defensins augment adaptive immune responses.
Article
Most bacteria that enter the bloodstream are taken up and eliminated within the liver. The specific mechanisms that underlie the role of the liver in the resolution of systemic bacterial infections remain to be determined. The vast majority of studies undertaken to date have focused on the function of resident tissue macrophages (Kupffer cells) that line the liver sinusoids. Indeed, it is often reported that Kupffer cells ingest and kill the bulk of organisms taken up by the liver. Recent studies indicate, however, that phagocytosis by Kupffer cells is not the principal mechanism by which organisms are eliminated. Rather, elimination depends on the complex interaction of Kupffer cells and bactericidal neutrophils that immigrate rapidly to the liver in response to infection. We discuss the critical role of neutrophil-Kupffer cell interaction in innate host defenses and, conceivably, the development and expression of adaptive immunity in the liver.
Article
Human neutrophil alpha-defensins (HNPs) are small, Cys-rich, cationic antimicrobial proteins. Stored in the azurophilic granules of neutrophils, they are released during phagocytosis to kill ingested foreign microbes through disruption of their cytoplasmic membrane. Recently, the three most abundant forms of human alpha-defensins, HNPs 1-3, have been implicated in suppressing HIV-1 infection in vivo, thereby exhibiting a potential therapeutic value in the treatment of AIDS. HNPs are synthesized as inactive precursors in vivo and require proteolytic removal of their inhibitory N-terminal pro-peptide for activation. Folding of HNPs 1-3 in vitro without the pro-peptide has been reported to be extremely difficult, which led to the hypothesis that the 45-residue anionic pro-peptide may assist proHNPs folding as an intramolecular chaperone interacting with the cationic C-terminal domain, a mechanism reminiscent of some bacterial serine proteases. Here we show that HNPs without the pro-region can fold productively with yields over 80% in the presence of 2 M urea and 25% N,N-dimethylformamide (DMF). Our finding demonstrates an efficient protocol for the production of large quantities of highly pure human alpha-defensins and is broadly applicable in folding aggregation-prone, Cys-rich proteins of both synthetic and recombinant origin.