Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
iScience ; 25(10): 105144, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36185363

ABSTRACT

Entosis is a cell competition process during which tumor cells engulf other tumor cells. It is initiated by metabolic stress or by loss of matrix adhesion, and it provides the winning cell with resources derived from the internalized cell. Using micropatterns as substrates for single cell migration, we find that the depletion of the cell adhesion receptor JAM-A strongly increases the rate of entosis in matrix-adherent cells. The activity of JAM-A in suppressing entosis depends on phosphorylation at Tyr280, which is a binding site for C-terminal Src kinase, and which we have previously found to regulate tumor cell motility and contact inhibition of locomotion (CIL). Loss of JAM-A triggers entosis in matrix-adherent cells but not matrix-deprived cells. Our findings strongly suggest that the increased motility and the perturbed CIL response after the depletion of JAM-A promote entotic cell engulfment, and they link a dysregulation of CIL to entosis in breast cancer cells.

2.
Sci Signal ; 15(751): eabm2449, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36099341

ABSTRACT

Intestinal epithelial cells absorb nutrients through the brush border, composed of dense arrays of highly ordered microvilli at their apical membranes. A protocadherin-based intermicrovillar adhesion complex localized at microvilli tips mediates microvilli packing and organization. Here, we identified a second adhesion complex localized at the proximal base region of microvilli. This complex contained the immunoglobulin superfamily member TMIGD1, which directly interacted with the microvillar scaffolding proteins EBP50 and E3KARP. Complex formation with EBP50 required the activation of EBP50 by the actin-binding protein ezrin and was enhanced by the dephosphorylation of Ser162 in the PDZ2 domain of EBP50 by the phosphatase PP1α. Binding of the EBP50-ezrin complex to TMIGD1 enhanced the dynamic turnover of EBP50 at microvilli. Enterocyte-specific inactivation of Tmigd1 in mice resulted in microvillar blebbing, loss of intermicrovillar adhesion, and perturbed brush border formation. Thus, we identified a second adhesion complex in microvilli and propose a mechanism that promotes microvillar formation and dynamics.


Subject(s)
Epithelial Cells , Intestines , Membrane Glycoproteins/metabolism , Animals , Cell Membrane/metabolism , Epithelial Cells/metabolism , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microvilli/metabolism
3.
J Cell Biol ; 221(4)2022 04 04.
Article in English | MEDLINE | ID: mdl-35293964

ABSTRACT

Contact inhibition of locomotion (CIL) is a process that regulates cell motility upon collision with other cells. Improper regulation of CIL has been implicated in cancer cell dissemination. Here, we identify the cell adhesion molecule JAM-A as a central regulator of CIL in tumor cells. JAM-A is part of a multimolecular signaling complex in which tetraspanins CD9 and CD81 link JAM-A to αvß5 integrin. JAM-A binds Csk and inhibits the activity of αvß5 integrin-associated Src. Loss of JAM-A results in increased activities of downstream effectors of Src, including Erk1/2, Abi1, and paxillin, as well as increased activity of Rac1 at cell-cell contact sites. As a consequence, JAM-A-depleted cells show increased motility, have a higher cell-matrix turnover, and fail to halt migration when colliding with other cells. We also find that proper regulation of CIL depends on αvß5 integrin engagement. Our findings identify a molecular mechanism that regulates CIL in tumor cells and have implications on tumor cell dissemination.


Subject(s)
Contact Inhibition , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Movement , Contact Inhibition/genetics , Receptors, Vitronectin , Tetraspanins
4.
PLoS Pathog ; 16(8): e1008775, 2020 08.
Article in English | MEDLINE | ID: mdl-32866218

ABSTRACT

Small RNA viruses only have a very limited coding capacity, thus most viral proteins have evolved to fulfill multiple functions. The highly conserved matrix protein 1 (M1) of influenza A viruses is a prime example for such a multifunctional protein, as it acts as a master regulator of virus replication whose different functions have to be tightly regulated. The underlying mechanisms, however, are still incompletely understood. Increasing evidence points towards an involvement of posttranslational modifications in the spatio-temporal regulation of M1 functions. Here, we analyzed the role of M1 tyrosine phosphorylation in genuine infection by using recombinant viruses expressing M1 phosphomutants. Presence of M1 Y132A led to significantly decreased viral replication compared to wildtype and M1 Y10F. Characterization of phosphorylation dynamics by mass spectrometry revealed the presence of Y132 phosphorylation in M1 incorporated into virions that is most likely mediated by membrane-associated Janus kinases late upon infection. Molecular dynamics simulations unraveled a potential phosphorylation-induced exposure of the positively charged linker domain between helices 4 and 5, supposably acting as interaction platform during viral assembly. Consistently, M1 Y132A showed a defect in lipid raft localization due to reduced interaction with viral HA protein resulting in a diminished structural stability of viral progeny and the formation of filamentous particles. Importantly, reduced M1-RNA binding affinity resulted in an inefficient viral genome incorporation and the production of non-infectious virions that interferes with virus pathogenicity in mice. This study advances our understanding of the importance of dynamic phosphorylation as a so far underestimated level of regulation of multifunctional viral proteins and emphasizes the potential feasibility of targeting posttranslational modifications of M1 as a novel antiviral intervention.


Subject(s)
Influenza A virus/metabolism , Mutation, Missense , Viral Matrix Proteins/metabolism , A549 Cells , Amino Acid Substitution , Animals , Dogs , Female , HEK293 Cells , Humans , Influenza A virus/genetics , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Transgenic , Phosphorylation , Viral Matrix Proteins/genetics
5.
Mol Imaging Biol ; 22(6): 1501-1510, 2020 12.
Article in English | MEDLINE | ID: mdl-32737655

ABSTRACT

PURPOSE: Extracellular vesicles, small vesicles carrying inter alia proteins, miRNA and RNA, are important mediators of intercellular communication. The purpose of this study was to assess the distribution of extracellular vesicles from highly malignant breast cancer and their subsequent effect on the immune cell infiltrate in target organs of metastasis. PROCEDURES: Extracellular vesicles were isolated from the tissue culture supernatant of highly malignant 4T1 breast cancer cells or the serum of healthy BALB/c mice. The purity of the isolate was verified by electron microscopy and western blotting. Extracellular vesicles were additionally subjected to proteome analysis. After labeling with the fluorescent dye DiR, extracellular vesicles were injected into healthy BALB/c mice and their in vivo distribution was assessed using fluorescence reflectance imaging (FRI). Following ex vivo imaging of the organs, lung tissue samples were analyzed for extracellular vesicle-mediated changes of myeloid cells and T cell numbers, using flow cytometry. Proteome analysis revealed major differences in the cargo of tumor cell-derived versus extracellular vesicles from healthy serum. RESULTS: In contrast to control extracellular vesicles, DiR-labeled extracellular vesicles from tumor cells preferentially accumulated in lung, liver, and spine. Subsequent flow cytometry of the immune cell composition of lung tissue samples revealed an increase of cytotoxic CD8+ T cells and a decrease of CD4+ T-helper cells as well as an increase in mature macrophages in response to tumor cell EV. CONCLUSIONS: In conclusion, distribution of tumor cell-derived extracellular vesicles follows a specific pattern and can be monitored, using dedicated imaging. Extracellular vesicles alter the immune cell composition in target organs of metastasis, using a specific proteome cargo.


Subject(s)
Extracellular Vesicles/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cell Line, Tumor , Extracellular Vesicles/ultrastructure , Female , Kinetics , Mice, Inbred BALB C , Neoplasm Metastasis , Proteome/metabolism , Tissue Distribution
6.
Clin Transl Gastroenterol ; 11(3): e00134, 2020 03.
Article in English | MEDLINE | ID: mdl-32352717

ABSTRACT

OBJECTIVES: In patients with inflammatory bowel disease (IBD), a treat-to-target treatment strategy requires tight monitoring of disease activity. Noninvasive biomarkers may help to monitor the intestinal disease activity. We demonstrated recently that peripheral microRNA (miR)-320a expression in mice follows the course of experimental colitis. The aim of this study was to evaluate the potential of miR-320a to monitor the disease activity in patients with IBD, to predict the course of disease, and to distinguish IBD from infectious colitis. METHODS: The miR-320a levels were prospectively assessed by quantitative real-time polymerase chain reaction analysis of peripheral blood samples from 40 patients with Crohn's disease (CD) and 37 patients with ulcerative colitis (UC) as well as from 19 healthy control individuals and 7 patients with infectious colitis. Disease activity was quantified by appropriate clinical disease indices and endoscopic scoring systems. RESULTS: When compared with healthy controls, miR-320a blood levels were significantly increased in patients with active CD and UC (16.1 ± 2.6 vs 2,573 ± 941; vs 434 ± 96; both P < 0.001) and patients with IBD in remission (316 ± 251 [CD] and 91 ± 29 [UC]; both P < 0.001). In patients with CD, miR-320a levels showed a strong correlation with the endoscopic disease activity (r = 0.76; P < 0.001). Similarly, in patients with UC, we detected a significantly enhanced miR-320a expression, which was highest in patients with severe endoscopic disease activity (eMayo = 0-1: 66 ± 16 vs eMayo = 2: 352 ± 102; vs eMayo = 3: 577 ± 206; both P < 0.001). Finally, miR-320a blood expression in patients with active CD and UC significantly increased compared with patients with infectious colitis (63 ± 13, P < 0.001). DISCUSSION: MiR-320a expression in peripheral blood from patients with IBD follows the clinical and endoscopic disease activities and may help to distinguish IBD from infectious colitis.


Subject(s)
Colitis, Ulcerative/diagnosis , Crohn Disease/diagnosis , MicroRNAs/blood , Adolescent , Adult , Biomarkers/blood , Case-Control Studies , Colitis, Ischemic/blood , Colitis, Ischemic/diagnosis , Colitis, Ischemic/microbiology , Colitis, Ulcerative/blood , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/diagnostic imaging , Colon/immunology , Colon/pathology , Colonoscopy , Crohn Disease/blood , Crohn Disease/immunology , Crohn Disease/pathology , Diagnosis, Differential , Enterocolitis, Pseudomembranous/blood , Enterocolitis, Pseudomembranous/diagnosis , Enterocolitis, Pseudomembranous/microbiology , Female , Healthy Volunteers , Humans , Intestinal Mucosa/diagnostic imaging , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Middle Aged , Severity of Illness Index , Young Adult
7.
J Virol ; 94(11)2020 05 18.
Article in English | MEDLINE | ID: mdl-32188731

ABSTRACT

Human papillomavirus 16 (HPV16), the leading cause of cervical cancer, exploits a novel endocytic pathway during host cell entry. This mechanism shares many requirements with macropinocytosis but differs in the mode of vesicle formation. Previous work indicated a role of the epidermal growth factor receptor (EGFR) in HPV16 endocytosis. However, the functional outcome of EGFR signaling and its downstream targets during HPV16 uptake are not well characterized. Here, we analyzed the functional importance of signal transduction via EGFR and its downstream effectors for endocytosis of HPV16. Our findings indicate two phases of EGFR signaling as follows: a-likely dispensable-transient activation with or shortly after cell binding and signaling required throughout the process of asynchronous internalization of HPV16. Interestingly, EGFR inhibition interfered with virus internalization and strongly reduced the number of endocytic pits, suggesting a role for EGFR signaling in the induction of HPV16 endocytosis. Moreover, we identified the Src-related kinase Abl2 as a novel regulator of virus uptake. Inhibition of Abl2 resulted in an accumulation of misshaped endocytic pits, indicating Abl2's importance for endocytic vesicle maturation. Since Abl2 rather than Src, a regulator of membrane ruffling during macropinocytosis, mediated downstream signaling of EGFR, we propose that the selective effector targeting downstream of EGFR determines whether HPV16 endocytosis or macropinocytosis is induced.IMPORTANCE Human papillomaviruses are small, nonenveloped DNA viruses that infect skin and mucosa. The so-called high-risk HPVs (e.g., HPV16, HPV18, HPV31) have transforming potential and are associated with various anogenital and oropharyngeal tumors. These viruses enter host cells by a novel endocytic pathway with unknown cellular function. To date, it is unclear how endocytic vesicle formation occurs mechanistically. Here, we addressed the role of epidermal growth factor receptor signaling, which has previously been implicated in HPV16 endocytosis and identified the kinase Abl2 as a novel regulator of virus uptake. Since other viruses, such as influenza A virus and lymphocytic choriomeningitis virus, possibly make use of related mechanisms, our findings shed light on fundamental strategies of virus entry and may in turn help to develop new host cell-targeted antiviral strategies.


Subject(s)
Endocytosis , Human papillomavirus 16/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Virus Internalization , Animals , ErbB Receptors/genetics , ErbB Receptors/metabolism , HeLa Cells , Human papillomavirus 16/genetics , Humans , Mice , Protein-Tyrosine Kinases/genetics
8.
J Virol ; 93(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30626687

ABSTRACT

Merkel cell polyomavirus (MCPyV) is a small, nonenveloped tumor virus associated with an aggressive form of skin cancer, Merkel cell carcinoma (MCC). MCPyV infections are highly prevalent in the human population, with MCPyV virions being continuously shed from human skin. However, the precise host cell tropism(s) of MCPyV remains unclear: MCPyV is able to replicate within a subset of dermal fibroblasts, but MCPyV DNA has also been detected in a variety of other tissues. However, MCPyV appears different from other polyomaviruses, as it requires sulfated polysaccharides, such as heparan sulfates and/or chondroitin sulfates, for initial attachment. Like other polyomaviruses, MCPyV engages sialic acid as a (co)receptor. To explore the infectious entry process of MCPyV, we analyzed the cell biological determinants of MCPyV entry into A549 cells, a highly transducible lung carcinoma cell line, in comparison to well-studied simian virus 40 and a number of other viruses. Our results indicate that MCPyV enters cells via caveolar/lipid raft-mediated endocytosis but not macropinocytosis, clathrin-mediated endocytosis, or glycosphingolipid-enriched carriers. The viruses were internalized in small endocytic pits that led the virus to endosomes and from there to the endoplasmic reticulum (ER). Similar to other polyomaviruses, trafficking required microtubular transport, acidification of endosomes, and a functional redox environment. To our surprise, the virus was found to acquire a membrane envelope within endosomes, a phenomenon not reported for other viruses. Only minor amounts of viruses reached the ER, while the majority was retained in endosomal compartments, suggesting that endosome-to-ER trafficking is a bottleneck during infectious entry.IMPORTANCE MCPyV is the first polyomavirus directly implicated in the development of an aggressive human cancer, Merkel cell carcinoma (MCC). Although MCPyV is constantly shed from healthy skin, the MCC incidence increases among aging and immunocompromised individuals. To date, the events connecting initial MCPyV infection and subsequent transformation still remain elusive. MCPyV differs from other known polyomaviruses concerning its cell tropism, entry receptor requirements, and infection kinetics. In this study, we examined the cellular requirements for endocytic entry as well as the subcellular localization of incoming virus particles. A thorough understanding of the determinants of the infectious entry pathway and the specific biological niche will benefit prevention of virus-derived cancers such as MCC.


Subject(s)
Merkel cell polyomavirus/pathogenicity , Polyomavirus Infections/virology , A549 Cells , Antigens, Viral, Tumor/metabolism , Carcinoma, Merkel Cell/virology , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Fibroblasts/virology , HEK293 Cells , HeLa Cells , Heparitin Sulfate/metabolism , Humans , Merkel cell polyomavirus/metabolism , N-Acetylneuraminic Acid/metabolism , Skin/virology , Skin Neoplasms/virology , Tumor Virus Infections/virology , Viral Tropism/physiology
9.
Int J Med Microbiol ; 308(8): 1027-1035, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30268774

ABSTRACT

The Gram-positive anaerobic bacterium Cutibacterium acnes is a commensal of the human skin, but also an opportunistic pathogen that contributes to the pathophysiology of the skin disease acne vulgaris. Moreover, C. acnes, in addition to other skin-colonizing bacteria such as S. epidermidis and S. aureus, is an emerging pathogen of implant-associated infections. Notably, C. acnes isolates exhibit marked heterogeneity and can be divided into at least 6 phylotypes by multilocus sequence typing. It is becoming increasingly evident that biofilm formation is a relevant factor for C. acnes virulence, but information on biofilm formation by diverse C. acnes isolates is limited. In this study we performed a first comparative analysis of 58 diverse skin- or implant-isolates covering all six C. acnes phylotypes to investigate biofilm formation dynamics, biofilm morphology and attachment properties to abiotic surfaces. The results presented herein suggest that biofilm formation correlates with the phylotype, rather than the anatomical isolation site. IA1 isolates, particularly SLST sub-types A1 and A2, showed highest biofilm amounts in the microtiter plate assays, followed by isolates of the IC, IA2 and II phylotypes. Microscopic evaluation revealed well-structured three-dimensional biofilms and relatively high adhesive properties to abiotic surfaces for phylotypes IA1, IA2 and IC. Representatives of phylotype III formed biofilms with comparable biomass, but with less defined structures, whereas IB as well as II isolates showed the least complex three-dimensional morphology. Proteinase K- and DNase I-treatment reduced attachment rates of all phylotypes, therefore, indicating that extracellular DNA and proteins are critical for adhesion to abiotic surfaces. Moreover, proteins seem to be pivotal structural biofilm components as mature biofilms of all phylotypes were proteinase K-sensitive, whereas the sensitivity to DNase I-treatment varied depending on the phylotype.


Subject(s)
Acne Vulgaris/microbiology , Biofilms/growth & development , Gram-Positive Bacterial Infections/microbiology , Propionibacteriaceae/growth & development , Skin/microbiology , Bacterial Adhesion/drug effects , Biofilms/drug effects , Deoxyribonuclease I/pharmacology , Endopeptidase K/pharmacology , Humans , Microbial Viability/drug effects , Microscopy, Fluorescence , Organic Chemicals/pharmacology , Propionibacteriaceae/drug effects , Propionibacteriaceae/isolation & purification
10.
Cell Microbiol ; 20(11): e12945, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30137651

ABSTRACT

The delivery of effector proteins into infected eukaryotic cells represents a key virulence feature of many microbial pathogens in order to derail essential cellular processes and effectively counter the host defence system. Although bacterial effectors are truly numerous and exhibit a wide range of biochemical activities, commonalities in terms of protein structure and function shared by many bacterial pathogens exist. Recent progress has shed light on a species-spanning family of bacterial effectors containing an LPX repeat motif as a subtype of the leucine-rich repeat superfamily, partially combined with a novel E3 ubiquitin ligase domain. This review highlights the immunomodulatory effects of LPX effector proteins, with particular emphasis on the exploitation of the host ubiquitin system.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Host-Pathogen Interactions/physiology , Amino Acid Motifs , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Humans , Salmonella/pathogenicity , Shigella/pathogenicity , Ubiquitin/metabolism
11.
Int J Med Microbiol ; 308(7): 872-881, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29936031

ABSTRACT

Microbial pathogens have developed intriguing molecular strategies to modulate and/or control host cell functions to ensure their own survival and replication. During this molecular interplay between microbes and their respective hosts especially secreted virulence factors play a major role. These factors not only include a plethora of cytotoxins but also sophisticated effector proteins targeting intracellular decision points leading to inhibition of defense responses - and/or even to cell death. To be effective, most of these secreted factors have to get across the cytoplasmic membrane and reach their targets in the cytoplasm. Apparently, pathogens use multiple mechanisms to deliver virulence factors to their cytoplasmic destination. Here, we exemplarily discuss the recently emerging scenario of parallel strategies for the intracellular deployment of toxins and effector proteins of Gram-negative pathogens with a special focus on pathogenic Escherichia coli. These pathogens employ various nanomachines such as the type III secretion system (T3SS), cell-penetrating effector proteins (CPE), and the wrapping of virulence factors in outer membrane vesicles (OMV) for protection and parallel delivery. As intracellular delivery remains a major problem in drug development, potential therapeutic applications based on these bacterial strategies will be briefly discussed.


Subject(s)
Bacterial Toxins/metabolism , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/transmission , Host-Pathogen Interactions/physiology , Type III Secretion Systems/physiology , Type IV Secretion Systems/physiology , Type VI Secretion Systems/physiology , Escherichia coli Infections/microbiology , Humans , Protein Transport/physiology , Virulence Factors/metabolism
12.
Int J Med Microbiol ; 308(3): 387-404, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29550166

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) subvert host cell signaling pathways by injecting effector proteins via a Type 3 Secretion System (T3SS). The T3SS-dependent EspB protein is a multi-functional effector protein, which contributes to adherence and translocator pore formation and after injection exhibits several intracellular activities. In addition, EspB is also secreted into the environment. Effects of secreted EspB have not been reported thus far. As a surrogate for secreted EspB we employed recombinant EspB (rEspB) derived from the prototype EPEC strain E2348/69 and investigated the interactions of the purified protein with different human epithelial and immune cells including monocytic THP-1 cells, macrophages, dendritic cells, U-937, epithelial T84, Caco-2, and HeLa cells. To assess whether these proteins might exert a cytotoxic effect we monitored the release of lactate dehydrogenase (LDH) as well as propidium iodide (PI) uptake. For comparison, we also investigated several homologs of EspB such as IpaD of Shigella, and SipC, SipD, SseB, and SseD of Salmonella as purified recombinant proteins. Interestingly, cytotoxicity was only observed in THP-1 cells and macrophages, whereas epithelial cells remained unaffected. Cell fractionation and immune fluorescence experiments showed that rEspB enters cells autonomously, which suggests that EspB might qualify as a novel cell-penetrating effector protein (CPE). Using specific organelle tracers and inhibitors of signaling pathways we found that rEspB destroys the mitochondrial membrane potential - an indication of programmed cell death induction in THP-1 cells. Here we show that EspB not only constitutes an essential part of the T3SS-nanomachine and contributes to the arsenal of injected effector proteins but, furthermore, that secreted (recombinant) EspB autonomously enters host cells and selectively induces cell death in immune cells.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Cell Death/genetics , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Monocytes/pathology , Bacterial Adhesion , Bacterial Proteins/genetics , Caco-2 Cells , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/pathogenicity , Epithelial Cells/microbiology , Epithelial Cells/pathology , HeLa Cells , Humans , L-Lactate Dehydrogenase/analysis , Monocytes/microbiology , Propidium/metabolism , Protein Transport , Salmonella/genetics , THP-1 Cells
13.
J Virol ; 92(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29593032

ABSTRACT

Human papillomavirus 16 (HPV16) is the leading cause of cervical cancer. For initial infection, HPV16 utilizes a novel endocytic pathway for host cell entry. Unique among viruses, uptake occurs asynchronously over a protracted period of time, with half-times between 9 and 12 h. To trigger endocytic uptake, the virus particles need to undergo a series of structural modifications after initial binding to heparan sulfate proteoglycans (HSPGs). These changes involve proteolytic cleavage of the major capsid protein L1 by kallikrein-8 (KLK8), exposure of the N terminus of the minor capsid protein L2 by cyclophilins, and cleavage of this N terminus by furin. Overall, the structural changes are thought to facilitate the engagement of an elusive secondary receptor for internalization. Here, we addressed whether structural changes are the rate-limiting steps during infectious internalization of HPV16 by using structurally primed HPV16 particles. Our findings indicate that the structural modifications mediated by cyclophilins and furin, which lead to exposure and cleavage, respectively, of the L2 N terminus contribute to the slow and asynchronous internalization kinetics, whereas conformational changes elicited by HSPG binding and KLK8 cleavage did not. However, these structural modifications accounted for only 30 to 50% of the delay in internalization. Therefore, we propose that limited internalization receptor availability for engagement of HPV16 causes slow and asynchronous internalization in addition to rate-limiting structural changes in the viral capsid.IMPORTANCE HPVs are the main cause of anogenital cancers. Their unique biology is linked to the differentiation program of skin or mucosa. Here, we analyzed another unique aspect of HPV infections using the prototype HPV16. After initial cell binding, HPVs display an unusually protracted residence time on the plasma membrane prior to asynchronous uptake. As viruses typically do not expose themselves to host immune sensing, we analyzed the underlying reasons for this unusual behavior. This study provides evidence that both extracellular structural modifications and possibly a limited availability of the internalization receptor contribute to the slow internalization process of the virus. These findings indicate that perhaps a unique niche for initial infection that could allow for rapid infection exists. In addition, our results may help to develop novel, preventive antiviral measures.


Subject(s)
Capsid/chemistry , Heparan Sulfate Proteoglycans/metabolism , Human papillomavirus 16/physiology , Endocytosis , HeLa Cells , Host-Pathogen Interactions , Human papillomavirus 16/metabolism , Humans , Kallikreins/metabolism , Protein Conformation , Virus Internalization
14.
Cell Mol Life Sci ; 75(12): 2273-2289, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29285573

ABSTRACT

Effector proteins are key virulence factors of pathogenic bacteria that target and subvert the functions of essential host defense mechanisms. Typically, these proteins are delivered into infected host cells via the type III secretion system (T3SS). Recently, however, several effector proteins have been found to enter host cells in a T3SS-independent manner thereby widening the potential range of these virulence factors. Prototypes of such bacteria-derived cell-penetrating effectors (CPEs) are the Yersinia enterocolitica-derived YopM as well as the Salmonella typhimurium effector SspH1. Here, we investigated specifically the group of bacterial LPX effector proteins comprising the Shigella IpaH proteins, which constitute a subtype of the leucine-rich repeat protein family and share significant homologies in sequence and structure. With particular emphasis on the Shigella-effector IpaH9.8, uptake into eukaryotic cell lines was shown. Recombinant IpaH9.8 (rIpaH9.8) is internalized via endocytic mechanisms and follows the endo-lysosomal pathway before escaping into the cytosol. The N-terminal alpha-helical domain of IpaH9.8 was identified as the protein transduction domain required for its CPE ability as well as for being able to deliver other proteinaceous cargo. rIpaH9.8 is functional as an ubiquitin E3 ligase and targets NEMO for poly-ubiquitination upon cell penetration. Strikingly, we could also detect other recombinant LPX effector proteins from Shigella and Salmonella intracellularly when applied to eukaryotic cells. In this study, we provide further evidence for the general concept of T3SS-independent translocation by identifying novel cell-penetrating features of these LPX effectors revealing an abundant species-spanning family of CPE.


Subject(s)
Bacterial Adhesion/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Conserved Sequence/genetics , Multigene Family , Protein Interaction Domains and Motifs/physiology , Virulence Factors/chemistry , Animals , Bacterial Proteins/metabolism , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Mice , Protein Interaction Domains and Motifs/genetics , RAW 264.7 Cells , Species Specificity , Virulence Factors/genetics , Virulence Factors/metabolism , Yersinia enterocolitica/chemistry , Yersinia enterocolitica/genetics , Yersinia enterocolitica/metabolism
15.
PLoS Pathog ; 13(5): e1006308, 2017 May.
Article in English | MEDLINE | ID: mdl-28464022

ABSTRACT

Incoming papillomaviruses (PVs) depend on mitotic nuclear envelope breakdown to gain initial access to the nucleus for viral transcription and replication. In our previous work, we hypothesized that the minor capsid protein L2 of PVs tethers the incoming vDNA to mitotic chromosomes to direct them into the nascent nuclei. To re-evaluate how dynamic L2 recruitment to cellular chromosomes occurs specifically during prometaphase, we developed a quantitative, microscopy-based assay for measuring the degree of chromosome recruitment of L2-EGFP. Analyzing various HPV16 L2 truncation-mutants revealed a central chromosome-binding region (CBR) of 147 amino acids that confers binding to mitotic chromosomes. Specific mutations of conserved motifs (IVAL286AAAA, RR302/5AA, and RTR313EEE) within the CBR interfered with chromosomal binding. Moreover, assembly-competent HPV16 containing the chromosome-binding deficient L2(RTR313EEE) or L2(IVAL286AAAA) were inhibited for infection despite their ability to be transported to intracellular compartments. Since vDNA and L2 were not associated with mitotic chromosomes either, the infectivity was likely impaired by a defect in tethering of the vDNA to mitotic chromosomes. However, L2 mutations that abrogated chromatin association also compromised translocation of L2 across membranes of intracellular organelles. Thus, chromatin recruitment of L2 may in itself be a requirement for successful penetration of the limiting membrane thereby linking both processes mechanistically. Furthermore, we demonstrate that the association of L2 with mitotic chromosomes is conserved among the alpha, beta, gamma, and iota genera of Papillomaviridae. However, different binding patterns point to a certain variance amongst the different genera. Overall, our data suggest a common strategy among various PVs, in which a central region of L2 mediates tethering of vDNA to mitotic chromosomes during cell division thereby coordinating membrane translocation and delivery to daughter nuclei.


Subject(s)
Capsid Proteins/metabolism , Genome, Viral/genetics , Human papillomavirus 16/genetics , Mitosis , Oncogene Proteins, Viral/metabolism , Biological Transport , Capsid Proteins/genetics , Cell Nucleus/metabolism , Cell Nucleus/virology , Chromatin/genetics , Chromosomes/genetics , DNA, Viral/genetics , DNA, Viral/metabolism , Genes, Reporter , Human papillomavirus 16/physiology , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/virology , Mutation , Oncogene Proteins, Viral/genetics , Virion
16.
Article in English | MEDLINE | ID: mdl-28451521

ABSTRACT

Effector proteins secreted by the type 3 secretion system (T3SS) of pathogenic bacteria have been shown to precisely modulate important signaling cascades of the host for the benefit of the pathogens. Among others, the non-LEE encoded T3SS effector protein NleC of enteropathogenic Escherichia coli (EPEC) is a Zn-dependent metalloprotease and suppresses innate immune responses by directly targeting the NF-κB signaling pathway. Many pathogenic bacteria release potent bacterial toxins of the A-B type, which-in contrast to the direct cytoplasmic injection of T3SS effector proteins-are released first into the environment. In this study, we found that NleC displays characteristics of bacterial A-B toxins, when applied to eukaryotic cells as a recombinant protein. Although lacking a B subunit, that typically mediates the uptake of toxins, recombinant NleC (rNleC) induces endocytosis via lipid rafts and follows the endosomal-lysosomal pathway. The conformation of rNleC is altered by low pH to facilitate its escape from acidified endosomes. This is reminiscent of the homologous A-B toxin AIP56 of the fish pathogen Photobacterium damselae piscicida (Phdp). The recombinant protease NleC is functional inside eukaryotic cells and cleaves p65 of the NF-κB pathway. Here, we describe the endocytic uptake mechanism of rNleC, characterize its intracellular trafficking and demonstrate that its specific activity of cleaving p65 requires activation of host cells e.g., by IL1ß. Further, we propose an evolutionary link between some T3SS effector proteins and bacterial toxins from apparently unrelated bacteria. In summary, these properties might suggest rNleC as an interesting candidate for future applications as a potential therapeutic against immune disorders.


Subject(s)
Enteropathogenic Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/toxicity , Recombinant Proteins , Transcription Factor RelA/metabolism , Type III Secretion Systems/metabolism , Amino Acid Sequence , Animals , Bacterial Toxins/metabolism , Endocytosis/drug effects , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/physiology , HeLa Cells , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Interleukin-1beta , Lysosomes/drug effects , NF-kappa B/metabolism , Photobacterium/metabolism , Protein Domains , Protein Structure, Secondary , Protein Transport , Sequence Alignment , Signal Transduction
17.
Virulence ; 8(7): 1124-1147, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28296562

ABSTRACT

Human-pathogenic Yersinia produce plasmid-encoded Yersinia outer proteins (Yops), which are necessary to down-regulate anti-bacterial responses that constrict bacterial survival in the host. These Yops are effectively translocated directly from the bacterial into the target cell cytosol by the type III secretion system (T3SS). Cell-penetrating peptides (CPPs) in contrast are characterized by their ability to autonomously cross cell membranes and to transport cargo - independent of additional translocation systems. The recent discovery of bacterial cell-penetrating effector proteins (CPEs) - with the prototype being the T3SS effector protein YopM - established a new class of autonomously translocating immunomodulatory proteins. CPEs represent a vast source of potential self-delivering, anti-inflammatory therapeutics. In this review, we give an update on the characteristic features of the plasmid-encoded Yops and, based on recent findings, propose the further development of these proteins for potential therapeutic applications as natural or artificial cell-penetrating forms of Yops might be of value as bacteria-derived biologics.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Yersinia Infections/immunology , Yersinia/immunology , Animals , Bacterial Outer Membrane Proteins/genetics , Humans , Type III Secretion Systems/genetics , Type III Secretion Systems/immunology , Yersinia/genetics , Yersinia Infections/microbiology
18.
PLoS Pathog ; 13(2): e1006159, 2017 02.
Article in English | MEDLINE | ID: mdl-28158302

ABSTRACT

Outer membrane vesicles (OMVs) are important tools in bacterial virulence but their role in the pathogenesis of infections caused by enterohemorrhagic Escherichia coli (EHEC) O157, the leading cause of life-threatening hemolytic uremic syndrome, is poorly understood. Using proteomics, electron and confocal laser scanning microscopy, immunoblotting, and bioassays, we investigated OMVs secreted by EHEC O157 clinical isolates for virulence factors cargoes, interactions with pathogenetically relevant human cells, and mechanisms of cell injury. We demonstrate that O157 OMVs carry a cocktail of key virulence factors of EHEC O157 including Shiga toxin 2a (Stx2a), cytolethal distending toxin V (CdtV), EHEC hemolysin, and flagellin. The toxins are internalized by cells via dynamin-dependent endocytosis of OMVs and differentially separate from vesicles during intracellular trafficking. Stx2a and CdtV-B, the DNase-like CdtV subunit, separate from OMVs in early endosomes. Stx2a is trafficked, in association with its receptor globotriaosylceramide within detergent-resistant membranes, to the Golgi complex and the endoplasmic reticulum from where the catalytic Stx2a A1 fragment is translocated to the cytosol. CdtV-B is, after its retrograde transport to the endoplasmic reticulum, translocated to the nucleus to reach DNA. CdtV-A and CdtV-C subunits remain OMV-associated and are sorted with OMVs to lysosomes. EHEC hemolysin separates from OMVs in lysosomes and targets mitochondria. The OMV-delivered CdtV-B causes cellular DNA damage, which activates DNA damage responses leading to G2 cell cycle arrest. The arrested cells ultimately die of apoptosis induced by Stx2a and CdtV via caspase-9 activation. By demonstrating that naturally secreted EHEC O157 OMVs carry and deliver into cells a cocktail of biologically active virulence factors, thereby causing cell death, and by performing first comprehensive analysis of intracellular trafficking of OMVs and OMV-delivered virulence factors, we provide new insights into the pathogenesis of EHEC O157 infections. Our data have implications for considering O157 OMVs as vaccine candidates.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Escherichia coli Infections/metabolism , Host-Pathogen Interactions/physiology , Virulence Factors/metabolism , Virulence/physiology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Escherichia coli O157 , Humans , Immunoblotting , Microscopy, Electron, Transmission , Protein Transport/physiology , Transport Vesicles/physiology
19.
Article in English | MEDLINE | ID: mdl-28096156

ABSTRACT

Commonly used antimicrobials show poor cellular uptake and often have limited access to intracellular targets, resulting in low antimicrobial activity against intracellular pathogens. An efficient delivery system to transport these drugs to the intracellular site of action is needed. Cell-penetrating peptides (CPPs) mediate the internalization of biologically active molecules into the cytoplasm. Here, we characterized two CPPs, α1H and α2H, derived from the Yersinia enterocolitica YopM effector protein. These CPPs, as well as Tat (trans-activator of transcription) from HIV-1, were used to deliver the antibiotic gentamicin to target intracellular bacteria. The YopM-derived CPPs penetrated different endothelial and epithelial cells to the same extent as Tat. CPPs were covalently conjugated to gentamicin, and CPP-gentamicin conjugates were used to target infected cells to kill multiple intracellular Gram-negative pathogenic bacteria, including Escherichia coli K1, Salmonella enterica serovar Typhimurium, and Shigella flexneri Taken together, CPPs show great potential as delivery vehicles for antimicrobial agents and may contribute to the generation of new therapeutic tools to treat infectious diseases caused by intracellular pathogens.


Subject(s)
Cell-Penetrating Peptides/chemistry , Gentamicins/chemistry , Gentamicins/pharmacology , Gram-Negative Bacteria/drug effects , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Escherichia coli/drug effects , Salmonella enterica/drug effects , Shigella flexneri/drug effects
20.
Trends Biotechnol ; 35(2): 109-120, 2017 02.
Article in English | MEDLINE | ID: mdl-27592802

ABSTRACT

Bacterial pathogens have developed intriguing virulence mechanisms, including several sophisticated nanomachines, for injecting effector proteins to manipulate host immune signaling pathways for their own benefit. Therefore, bacterial genomes harbor a wealth of information about how to manipulate the defense systems of the host. Current understanding addresses virulence mechanisms mostly as targets for antimicrobials. We propose a change of paradigm by exploiting bacterial effectors not as targets but as tools for the directed manipulation of host signaling - for the benefit of the host. Recently, effector proteins have been identified that autonomously translocate into host cells, representing a novel class of cell-penetrating peptides (CPPs) or effectors (CPEs). Moreover, autonomous cell penetration overcomes a major hurdle in pharmacology by transducing specific therapeutic agents to intracellular targets.


Subject(s)
Bacteria/immunology , Bacterial Proteins/immunology , Cell Physiological Phenomena/immunology , Cell-Penetrating Peptides/immunology , Host-Parasite Interactions/immunology , Immunity, Innate/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...