1
|
Van Staden ADP, Visser JG, Powrie YSL, Smith C. Harnessing Microbial Effectors for Macrophage-Mediated Drug Delivery. ACS OMEGA 2024; 9:18260-18272. [PMID: 38680365 PMCID: PMC11044259 DOI: 10.1021/acsomega.3c10519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/21/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024]
Abstract
Macrophage-based drug delivery systems are promising, but their development is still in its infancy, with many limitations remaining to be addressed. Our aim was to design a system harnessing microbial effectors to facilitate controlled drug cargo expulsion from macrophages to enable the use of more toxic drugs without adding to the risk of off-target detrimental effects. The pore forming and actin polymerizing Listeria monocytogenes effectors listeriolysin-O (LLO) and actin assembly-inducing protein (ActA) were synthesized using a novel green fluorescent protein (GFP)-linked heterologous expression system. These effectors were coated onto polystyrene beads to generate "synthetic cargo" before loading into primary M1 macrophages. Bead uptake and release from macrophages were evaluated by using high-throughput quantitative imaging flow cytometry and confocal microscopy. In vitro results confirmed appropriate activity of synthesized effectors. Coating of these effector proteins onto polystyrene beads (simulated drug cargo) resulted in changes in cellular morphology, bead content, and intracellular bead localization, which may support an interpretation of the induced release of these beads from the cells. This forms the basis for further investigation to fully elucidate any potential release mechanisms. Bacterial effectors ActA and LLO successfully effectuated actin polarization and protrusions from cell membranes similar to those seen in cells infected with Listeria spp., illustrating the potential of using these effectors and production methods for the development of an endogenous drug delivery system capable of low-risk, targeted release of high potency drugs.
Collapse
Affiliation(s)
- Anton Du Preez Van Staden
- Department
of Microbiology, Science Faculty, Stellenbosch
University, Stellenbosch 7600, South Africa
- Experimental
Medicine Research Group, Department of Medicine, Faculty of Medicine
and Health Sciences, Stellenbosch University, Parow 7505, South Africa
| | - Johan G. Visser
- Department
of Physiological Sciences, Science Faculty, Stellenbosch University, Stellenbosch 7602, South Africa
| | - Yigael S. L. Powrie
- Experimental
Medicine Research Group, Department of Medicine, Faculty of Medicine
and Health Sciences, Stellenbosch University, Parow 7505, South Africa
- Division
of Neurosurgery, University of Cape Twon, Cape Town 7925, South Africa
| | - Carine Smith
- Experimental
Medicine Research Group, Department of Medicine, Faculty of Medicine
and Health Sciences, Stellenbosch University, Parow 7505, South Africa
| |
Collapse
|
2
|
Quaresma TC, de Aguiar Valentim L, de Sousa JR, de Souza Aarão TL, Fuzii HT, Duarte MIS, de Souza J, Quaresma JAS. Immunohistochemical Characterization of M1, M2, and M4 Macrophages in Leprosy Skin Lesions. Pathogens 2023; 12:1225. [PMID: 37887741 PMCID: PMC10610015 DOI: 10.3390/pathogens12101225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Mycobacterium leprae is the etiological agent of leprosy. Macrophages (Mφs) are key players involved in the pathogenesis of leprosy. In this study, immunohistochemical analysis was performed to examine the phenotype of Mφ subpopulations, namely M1, M2, and M4, in the skin lesions of patients diagnosed with leprosy. Based on the database of treatment-naïve patients treated between 2015 and 2019 at the Department of Dermatology of the University of the State of Pará, Belém, routine clinical screening samples were identified. The monolabeling protocol was used for M1 macrophages (iNOS, IL-6, TNF-α) and M2 macrophages (IL-10, IL-13, CD163, Arginase 1, TGF-β, FGFb), and the double-labeling protocol was used for M4 macrophages (IL-6, MMP7, MRP8, TNF-α e CD68). To confirm the M4 macrophage lineage, double labeling of the monoclonal antibodies CD68 and MRP8 was also performed. Our results demonstrated a statistically significant difference for the M1 phenotype among the Virchowian (VV) (4.5 ± 1.3, p < 0.0001), Borderline (1.6 ± 0.4, p < 0.0001), and tuberculoid (TT) (12.5 ± 1.8, p < 0.0001) clinical forms of leprosy. Additionally, the M2 phenotype showed a statistically significant difference among the VV (12.5 ± 2.3, p < 0.0001), Borderline (1.3 ± 0.2, p < 0.0001), and TT (3.2 ± 0.7, p < 0.0001) forms. For the M4 phenotype, a statistically significant difference was observed in the VV (9.8 ± 1.7, p < 0.0001), Borderline (1.2 ± 0.2, p < 0.0001), and TT (2.6 ± 0.7, p < 0.0001) forms. A significant correlation was observed between the VV M1 and M4 (r = 0.8712; p = 0.0000) and between the VV M2 × TT M1 (r = 0.834; p = 0.0002) phenotypes. The M1 Mφs constituted the predominant Mφ subpopulation in the TT and Borderline forms of leprosy, whereas the M2 Mφs showed increased immunoexpression and M4 was the predominant Mφ phenotype in VV leprosy. These results confirm the relationship of the Mφ profile with chronic pathological processes of the inflammatory response in leprosy.
Collapse
Affiliation(s)
- Tatiane Costa Quaresma
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Lívia de Aguiar Valentim
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Jorge Rodrigues de Sousa
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Tinara Leila de Souza Aarão
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
- School of Medicine, Federal University of Para-UFPA, Altamira 68440-000, Brazil
| | - Hellen Thais Fuzii
- Health Department, Tropical Medicine Center, Federal University of Para-NMT-UFPA, Belem 66055-240, Brazil
| | | | - Juarez de Souza
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Juarez Antônio Simões Quaresma
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
- School of Medicine, Federal University of Para-UFPA, Altamira 68440-000, Brazil
- Health Department, Tropical Medicine Center, Federal University of Para-NMT-UFPA, Belem 66055-240, Brazil
- School of Medicine, Sao Paulo University, Sao Paulo 01246-903, Brazil
| |
Collapse
|
3
|
Innate immune activation and modulatory factors of Helicobacter pylori towards phagocytic and nonphagocytic cells. Curr Opin Immunol 2023; 82:102301. [PMID: 36933362 DOI: 10.1016/j.coi.2023.102301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/17/2023] [Indexed: 03/18/2023]
Abstract
Helicobacter pylori is an intriguing obligate host-associated human pathogen with a specific host interaction biology, which has been shaped by thousands of years of host-pathogen coevolution. Molecular mechanisms of interaction of H. pylori with the local immune cells in the human system are less well defined than epithelial cell interactions, although various myeloid cells, including neutrophils and other phagocytes, are locally present or attracted to the sites of infection and interact with H. pylori. We have recently addressed the question of novel bacterial innate immune stimuli, including bacterial cell envelope metabolites, that can activate and modulate cell responses via the H. pylori Cag type IV secretion system. This review article gives an overview of what is currently known about the interaction modes and mechanisms of H. pylori with diverse human cell types, with a focus on bacterial metabolites and cells of the myeloid lineage including phagocytic and antigen-presenting cells.
Collapse
|
4
|
Single-phagosome imaging reveals that homotypic fusion impairs phagosome degradative function. Biophys J 2022; 121:459-469. [PMID: 34968424 PMCID: PMC8822610 DOI: 10.1016/j.bpj.2021.12.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/01/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023] Open
Abstract
Immune cells degrade internalized pathogens in vesicle compartments called phagosomes. Many intracellular bacteria induce homotypic phagosome fusion to survive in host cells, but the fusion interaction between phagosomes and its consequence for phagosome function have scarcely been studied. Here, we characterize homotypic fusion between phagosomes in macrophages and identify how such interactions impact the degradative capacity of phagosomes. By developing a series of particle sensors for measuring biochemical changes of single phagosomes, we show that phagosomes undergo stable fusion, transient "kiss-and-run" fusion, or both in succession. Super-resolution three-dimensional fluorescence microscopy revealed that stably fused phagosomes are connected by membrane "necks" with submicron-sized fusion pores. Furthermore, we demonstrate that, after stable fusion, phagosomes have leaky membranes and thereby impaired degradative functions. Our findings, based on phagosomes that contain synthetic particles, illustrate that homotypic fusion is not exclusive to phagosomes that encapsulate pathogens, as previously believed. The physical process of homotypic fusion is alone sufficient to perturb the degradative functions of phagosomes.
Collapse
|
5
|
Coletta S, Battaggia G, Della Bella C, Furlani M, Hauke M, Faass L, D'Elios MM, Josenhans C, de Bernard M. ADP-heptose enables Helicobacter pylori to exploit macrophages as a survival niche by suppressing antigen-presenting HLA-II expression. FEBS Lett 2021; 595:2160-2168. [PMID: 34216493 DOI: 10.1002/1873-3468.14156] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022]
Abstract
The persistence of Helicobacter pylori in the human gastric mucosa implies that the immune response fails to clear the infection. We found that H. pylori compromises the antigen presentation ability of macrophages, because of the decline of the presenting molecules HLA-II. Here, we reveal that the main bacterial factor responsible for this effect is ADP-heptose, an intermediate metabolite in the biosynthetic pathway of lipopolysaccharide (LPS) that elicits a pro-inflammatory response in gastric epithelial cells. In macrophages, it upregulates the expression of miR146b which, in turn, would downmodulate CIITA, the master regulator for HLA-II genes. Hence, H. pylori, utilizing ADP-heptose, exploits a specific arm of macrophage response to establish its survival niche in the face of the immune defense elicited in the gastric mucosa.
Collapse
Affiliation(s)
- Sara Coletta
- Department of Biology, University of Padova, Italy
| | | | - Chiara Della Bella
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | | | - Martina Hauke
- Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, LMU Munich, Germany
| | - Larissa Faass
- Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, LMU Munich, Germany
| | - Mario M D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Christine Josenhans
- Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, LMU Munich, Germany
| | | |
Collapse
|
6
|
Sharma G, Sharma M, Sood R, Neelamraju J, Lakshmi SG, Madempudi RS, Rishi P, Kaur IP. Self-preserving gelatin emulgel containing whole cell probiotic for topical use: preclinical safety, efficacy, and germination studies. Expert Opin Drug Deliv 2021; 18:1777-1789. [PMID: 34176401 DOI: 10.1080/17425247.2021.1947239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Dermal disorders, owing to disruption of skin-microflora balance can be served by direct application of probiotics. However, there are few topical whole probiotic products in market because of (i) loss of viability during manufacturing and storage(ii) inadequate germination and retention on skin. Presently we report a novel (IPA 201811010395) emulgel incorporatingBacillus coagulans (Unique IS-2) for possible topical use. METHODS Developed emulgel was characterized for particle size, texture, rheology, morphology, water activity, self-preservation, safety, and stability. RESULTS We successfully incorporated 97 ± 5% (1.7×108CFU/g) Bacillus coagulans in honeycomb network of gelatin nanoparticles (≈600 nm). Maintenance of CFU at 30 ± 2°C, 65 ± 5% RH for 3 months confirmed viability of incorporated probiotic. Low water-activity (0.66-0.732aw) and challenge test (0.05-0.5% viability) confirmed its self-preserving nature. Early initiation (6 h) and complete (24 h) spore germination was evident onrabbit skin. No cytotoxicity, dermal irritation or translocation established its safety. Faster wound closure and reduced oxidative stress (LPO, catalase, SOD, glutathione reductase) in comparison to Soframycin® (1%w/w Framycetin) was observed in excision wound in mice. CONCLUSIONS A whole cell probiotic formulation that is self-preserving, maintains probiotic viability, guarantees germination, and has wound healing properties was successfully formulated.
Collapse
Affiliation(s)
- Garima Sharma
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Manuhaar Sharma
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Rishav Sood
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | | | | | | | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Indu Pal Kaur
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
7
|
Wen J, Chen C, Luo M, Liu X, Guo J, Wei T, Gu X, Gu S, Ning Y, Li Y. Notch Signaling Ligand Jagged1 Enhances Macrophage-Mediated Response to Helicobacter pylori. Front Microbiol 2021; 12:692832. [PMID: 34305857 PMCID: PMC8297740 DOI: 10.3389/fmicb.2021.692832] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori (H. pylori) is one of the gram-negative bacteria that mainly colonize the stomach mucosa and cause many gastrointestinal diseases, such as gastritis, peptic ulcer, and gastric cancer. Macrophages play a key role in eradicating H. pylori. Recent data have shown that Notch signaling could modulate the activation and bactericidal activities of macrophages. However, the role of Notch signaling in macrophages against H. pylori remains unclear. In the present study, in the co-culture model of macrophages with H. pylori, the inhibition of Notch signaling using γ-secretase decreased the expression of inducible nitric oxide synthase (iNOS) and its product, nitric oxide (NO), and downregulated the secretion of pro-inflammatory cytokine and attenuated phagocytosis and bactericidal activities of macrophages to H. pylori. Furthermore, we identified that Jagged1, one of Notch signaling ligands, was both upregulated in mRNA and protein level in activated macrophages induced by H. pylori. Clinical specimens showed that the number of Jagged1+ macrophages in the stomach mucosa from H. pylori-infected patients was significantly higher than that in healthy control. The overexpression of Jagged1 promoted bactericidal activities of macrophages against H. pylori and siRNA-Jagged1 presented the opposite effect. Besides, the addition of exogenous rJagged1 facilitated the pro-inflammatory mediators of macrophages against H. pylori, but the treatment of anti-Jagged1 neutralizing antibody attenuated it. Taken together, these results suggest that Jagged1 is a promoting molecule for macrophages against H. pylori, which will provide insight for exploring Jagged1 as a novel therapeutic target for the control of H. pylori infection.
Collapse
Affiliation(s)
- Junjie Wen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Chuxi Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Meiqun Luo
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaocong Liu
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Jiading Guo
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Tingting Wei
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Xinyi Gu
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Sinan Gu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yunshan Ning
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yan Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Arismendi Sosa AC, Salinas Ibáñez AG, Pérez Chaca MV, Penissi AB, Gómez NN, Vega AE. Inflammatory response induced by Helicobacter pylori infection in lung. Microb Pathog 2020; 142:104103. [PMID: 32112810 DOI: 10.1016/j.micpath.2020.104103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
Helicobacter pylori is a microorganism that in the last years has been associated with extragastric disorders such as respiratory diseases, however, its impact on lung is partially understood. The aim of this work was to study infection impact of H. pylori on the inflammatory markers expression at the pulmonary level using an animal model. Infection was performed by BALB/c wild type (WT) mice orotracheal instillation with 20 μl of 1 × 108H. pylori reference strain suspension once per day throughout 3 days. Inflammatory response was evaluated at 3, 7, 14, 21 and 30 days post infection. Lung was aseptically removed and pulmonary edema index values showed a significant change at 30 days of infection. Hematoxylin-Eosin (H-E) stain allowed to visualizing H. pylori presence in lung samples at 3 days of infection near the phagocytic cells or in the alveoli lumen. Bronchoalveolar lavage (BAL) was used for inflammatory response evaluation. Lactate dehydrogenase values showed a gradual increase in infected animals along infection time. Protein concentrations in mg/ml from BAL increased significantly at 7 days in infected animals. Macrophages viability obtained from BAL, decreased at the first moment of infection, maintaining constant values along contamination time. Results obtained demonstrate an inflammatory response in lung after orotracheal H. pylori infection and suggest that the pathogenic mechanism is strongly evidenced by tissue damage, endothelial dysfunction inflammatory mediators and markers expression at the pulmonary level.
Collapse
Affiliation(s)
- A C Arismendi Sosa
- Área Microbiología e Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina.
| | - A G Salinas Ibáñez
- Área Microbiología e Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - M V Pérez Chaca
- Área de Morfología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - A B Penissi
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - N N Gómez
- Área de Morfología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis, Argentina
| | - A E Vega
- Área Microbiología e Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| |
Collapse
|
9
|
Codolo G, Toffoletto M, Chemello F, Coletta S, Soler Teixidor G, Battaggia G, Munari G, Fassan M, Cagnin S, de Bernard M. Helicobacter pylori Dampens HLA-II Expression on Macrophages via the Up-Regulation of miRNAs Targeting CIITA. Front Immunol 2020; 10:2923. [PMID: 31969878 PMCID: PMC6960189 DOI: 10.3389/fimmu.2019.02923] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/28/2019] [Indexed: 12/17/2022] Open
Abstract
Macrophages have a major role in infectious and inflammatory diseases, and the available data suggest that Helicobacter pylori persistence can be explained in part by the failure of the bacterium to be killed by professional phagocytes. Macrophages are cells ready to kill the engulfed pathogen, through oxygen-dependent and -independent mechanisms; however, their killing potential can be further augmented by the intervention of T helper (Th) cells upon the specific recognition of human leukocyte antigen (HLA)-II–peptide complexes on the surface of the phagocytic cells. As it pertains to H. pylori, the bacterium is engulfed by macrophages, but it interferes with the phagosome maturation process leading to phagosomes with an altered degradative capacity, and to megasomes, wherein H. pylori resists killing. We recently showed that macrophages infected with H. pylori strongly reduce the expression of HLA-II molecules on the plasma membrane and this compromises the bacterial antigen presentation to Th lymphocytes. In this work, we demonstrate that H. pylori hampers HLA-II expression in macrophages, activated or non-activated by IFN-γ, by down-regulating the expression of the class II major histocompatibility complex transactivator (CIITA), the “master control factor” for the expression of HLA class II genes. We provided evidence that this effect relies on the up-regulation of let-7f-5p, let-7i-5p, miR-146b-5p, and -185-5p targeting CIITA. MiRNA expression analysis performed on biopsies from H. pylori-infected patients confirmed the up-regulation of let-7i-5p, miR-146b-5p, and -185-5p in gastritis, in pre-invasive lesions, and in gastric cancer. Taken together, our results suggest that specific miRNAs may be directly involved in the H. pylori infection persistence and may contribute to confer the risk of developing gastric neoplasia in infected patients.
Collapse
Affiliation(s)
- Gaia Codolo
- Department of Biology, University of Padua, Padua, Italy
| | | | - Francesco Chemello
- Department of Biology, University of Padua, Padua, Italy.,CRIBI Biotechnology Center, University of Padua, Padua, Italy
| | - Sara Coletta
- Department of Biology, University of Padua, Padua, Italy
| | | | | | - Giada Munari
- Istituto Oncologico Veneto (IRCCS), Padua, Italy
| | - Matteo Fassan
- Department of Medicine, University of Padua, Padua, Italy
| | - Stefano Cagnin
- Department of Biology, University of Padua, Padua, Italy.,CRIBI Biotechnology Center, University of Padua, Padua, Italy.,CIR-Myo Myology Center, University of Padua, Padua, Italy
| | | |
Collapse
|
10
|
Lian DW, Xu YF, Deng QH, Lin XM, Huang B, Xian SX, Huang P. Effect of patchouli alcohol on macrophage mediated Helicobacter pylori digestion based on intracellular urease inhibition. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 65:153097. [PMID: 31568921 DOI: 10.1016/j.phymed.2019.153097] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/18/2019] [Accepted: 09/24/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Helicobacter pylori infects almost half of the world population and is listed as a type I carcinoma factor since 1994. Pogostemon cablin (Blanco) Benth. (Labiatae) has been used to treat gastro-intestinal diseases for thousands of years in many east Asian countries, and the key ingredient, patchouli alcohol (PA), has been observed to exert anti-H. pylori and anti-urease activities. PURPOSE We investigated the effect of PA on H. pylori urease and its subsequent influence on macrophage phagosome maturation and function. METHODS In H. pylori experiment, the berthelot method and pH shock assay were adopted to evaluate the effect of PA on extracellular and intracellular H. pylori urease. And then, Q-PCR and Western blot were carried out to analyze the alterations in the expression of urease-related genes and proteins after PA treatment. In the H. pylori and macrophage cell (RAW264.7) co-culture experiment, the effects of PA on H. pylori-induced phagocytosis and intracellular killing of RAW264.7 were investigated using gentamycin protection assay, and the underlying mechanism was explored by immunofluorescence. RESULTS PA at 25 and 50 μM inhibited intracellular H. pylori urease activity but not isolated urease by down-regulating the gene expression levels of ureB, ureE, ureI and nixA and reducing the protein expression level of UreB, thereby inhibiting the acid resistance of H. pylori. PA also recovered the function of macrophage bacterial digestion, and prior treatment with ammonium chloride inhibited the efficacy of PA. CONCLUSION PA suppressed intracellular H. pylori urease function and maturation, which increased macrophage digestion ability.
Collapse
Affiliation(s)
- D W Lian
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, PR China; Lingnan Medical Research Center, Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, PR China; Postdoctoral Research Station of Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Y F Xu
- Shenzhen Traditional Chinese Medicine Hospital, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, PR China; Postdoctoral Research Station of Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Q H Deng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - X M Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - B Huang
- Shenzhen Traditional Chinese Medicine Hospital, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, PR China
| | - S X Xian
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, PR China; Lingnan Medical Research Center, Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, PR China.
| | - P Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| |
Collapse
|
11
|
Blaser N, Backert S, Pachathundikandi SK. Immune Cell Signaling by Helicobacter pylori: Impact on Gastric Pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1149:77-106. [PMID: 31049845 DOI: 10.1007/5584_2019_360] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori represents a highly successful colonizer of the human stomach. Infections with this Gram-negative bacterium can persist lifelong, and although in the majority of cases colonization is asymptomatic, it can trigger pathologies ranging from chronic gastritis and peptic ulceration to gastric cancer. The interaction of the bacteria with the human host modulates immune responses in different ways to enable bacterial survival and persistence. H. pylori uses various pathogenicity-associated factors such as VacA, NapA, CGT, GGT, lipopolysaccharide, peptidoglycan, heptose 1,7-bisphosphate, ADP-heptose, cholesterol glucosides, urease and a type IV secretion system for controlling immune signaling and cellular functions. It appears that H. pylori manipulates multiple extracellular immune receptors such as integrin-β2 (CD18), EGFR, CD74, CD300E, DC-SIGN, MINCLE, TRPM2, T-cell and Toll-like receptors as well as a number of intracellular receptors including NLRP3, NOD1, NOD2, TIFA and ALPK1. Consequently, downstream signaling pathways are hijacked, inducing tolerogenic dendritic cells, inhibiting effector T cell responses and changing the gastrointestinal microbiota. Here, we discuss in detail the interplay of bacterial factors with multiple immuno-regulatory cells and summarize the main immune evasion and persistence strategies employed by H. pylori.
Collapse
Affiliation(s)
- Nicole Blaser
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Steffen Backert
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Suneesh Kumar Pachathundikandi
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
12
|
de Sousa JR, Da Costa Vasconcelos PF, Quaresma JAS. Functional aspects, phenotypic heterogeneity, and tissue immune response of macrophages in infectious diseases. Infect Drug Resist 2019; 12:2589-2611. [PMID: 31686866 PMCID: PMC6709804 DOI: 10.2147/idr.s208576] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/05/2019] [Indexed: 12/13/2022] Open
Abstract
Macrophages are a functionally heterogeneous group of cells with specialized functions depending not only on their subgroup but also on the function of the organ or tissue in which the cells are located. The concept of macrophage phenotypic heterogeneity has been investigated since the 1980s, and more recent studies have identified a diverse spectrum of phenotypic subpopulations. Several types of macrophages play a central role in the response to infectious agents and, along with other components of the immune system, determine the clinical outcome of major infectious diseases. Here, we review the functions of various macrophage phenotypic subpopulations, the concept of macrophage polarization, and the influence of these cells on the evolution of infections. In addition, we emphasize their role in the immune response in vivo and in situ, as well as the molecular effectors and signaling mechanisms used by these cells. Furthermore, we highlight the mechanisms of immune evasion triggered by infectious agents to counter the actions of macrophages and their consequences. Our aim here is to provide an overview of the role of macrophages in the pathogenesis of critical transmissible diseases and discuss how elucidation of this relationship could enhance our understanding of the host-pathogen association in organ-specific immune responses.
Collapse
Affiliation(s)
- Jorge Rodrigues de Sousa
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
| | - Pedro Fernando Da Costa Vasconcelos
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
| | - Juarez Antonio Simões Quaresma
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
- School of Medicine, São Paulo University, São Paulo, SP, Brazil
| |
Collapse
|
13
|
Pagliari M, Munari F, Toffoletto M, Lonardi S, Chemello F, Codolo G, Millino C, Della Bella C, Pacchioni B, Vermi W, Fassan M, de Bernard M, Cagnin S. Helicobacter pylori Affects the Antigen Presentation Activity of Macrophages Modulating the Expression of the Immune Receptor CD300E through miR-4270. Front Immunol 2017; 8:1288. [PMID: 29085364 PMCID: PMC5649134 DOI: 10.3389/fimmu.2017.01288] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/25/2017] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori (Hp) is a Gram-negative bacterium that infects the human gastric mucosa, leading to chronic inflammation. If not eradicated with antibiotic treatment, the bacterium persists in the human stomach for decades increasing the risk to develop chronic gastritis, gastroduodenal ulcer, and gastric adenocarcinoma. The lifelong persistence of Hp in the human stomach suggests that the host response fails to clear the infection. It has been recently shown that during Hp infection phagocytic cells promote high Hp loads rather than contributing to bacterial clearance. Within these cells Hp survives in "megasomes," large structures arising from homotypic fusion of phagosomes, but the mechanism that Hp employs to avoid phagocytic killing is not completely understood. Here, we show that Hp infection induces the downregulation of specific microRNAs involved in the regulation of transcripts codifying for inflammatory proteins. miR-4270 targets the most upregulated gene: the immune receptor CD300E, whose expression is strictly dependent on Hp infection. CD300E engagement enhances the pro-inflammatory potential of macrophages, but in parallel it affects their ability to express and expose MHC class II molecules on the plasma membrane, without altering phagocytosis. This effect compromises the possibility for effector T cells to recognize and activate the killing potential of macrophages, which, in turn would become a survival niche for the bacterium. Taken together, our data add another piece to the complicate puzzle represented by the long-life coexistence between Hp and the human host and contribute with new insights toward understanding the regulation and function of the immune receptor CD300E.
Collapse
Affiliation(s)
| | - Fabio Munari
- Department of Biomedical Sciences, University of Padua, Venetian Institute of Molecular Medicine (VIMM), Padua, Italy
| | | | - Silvia Lonardi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - Francesco Chemello
- Department of Biology, University of Padua, Padua, Italy.,CRIBI Biotechnology Center, University of Padua, Padua, Italy
| | - Gaia Codolo
- Department of Biology, University of Padua, Padua, Italy
| | - Caterina Millino
- Department of Biology, University of Padua, Padua, Italy.,CRIBI Biotechnology Center, University of Padua, Padua, Italy
| | | | - Beniamina Pacchioni
- Department of Biology, University of Padua, Padua, Italy.,CRIBI Biotechnology Center, University of Padua, Padua, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - Matteo Fassan
- Department of Medicine, University of Padua, Padua, Italy
| | | | - Stefano Cagnin
- Department of Biology, University of Padua, Padua, Italy.,CRIBI Biotechnology Center, University of Padua, Padua, Italy
| |
Collapse
|
14
|
Galectin-3 Plays an Important Role in Innate Immunity to Gastric Infection by Helicobacter pylori. Infect Immun 2016; 84:1184-1193. [PMID: 26857579 DOI: 10.1128/iai.01299-15] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/02/2016] [Indexed: 02/06/2023] Open
Abstract
We studied the role of galectin-3 (Gal3) in gastric infection by Helicobacter pylori We first demonstrated that Gal3 was selectively expressed by gastric surface epithelial cells and abundantly secreted into the surface mucus layer. We next inoculated H. pylori Sydney strain 1 into wild-type (WT) and Gal3-deficient mice using a stomach tube. At 2 weeks postinoculation, the bacterial cells were mostly trapped within the surface mucus layer in WT mice. In sharp contrast, they infiltrated deep into the gastric glands in Gal3-deficient mice. Bacterial loads in the gastric tissues were also much higher in Gal3-deficient mice than in WT mice. At 6 months postinoculation,H. pylori had successfully colonized within the gastric glands of both WT and Gal3-deficient mice, although the bacterial loads were still higher in the latter. Furthermore, large lymphoid clusters mostly consisting of B cells were frequently observed in the gastric submucosa of Gal3-deficient mice.In vitro, peritoneal macrophages from Gal3-deficient mice were inefficient in killing engulfed H. pylori Furthermore, recombinant Gal3 not only induced rapid aggregation of H. pylori but also exerted a potent bactericidal effect on H. pylori as revealed by propidium iodide uptake and a morphological shift from spiral to coccoid form. However, a minor fraction of bacterial cells, probably transient phase variants of Gal3-binding sugar moieties, escaped killing by Gal3. Collectively, our data demonstrate that Gal3 plays an important role in innate immunity to infection and colonization of H. pylori.
Collapse
|
15
|
Kronsteiner B, Bassaganya-Riera J, Philipson C, Viladomiu M, Carbo A, Abedi V, Hontecillas R. Systems-wide analyses of mucosal immune responses to Helicobacter pylori at the interface between pathogenicity and symbiosis. Gut Microbes 2016; 7:3-21. [PMID: 26939848 PMCID: PMC4856448 DOI: 10.1080/19490976.2015.1116673] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/29/2015] [Accepted: 10/31/2015] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori is the dominant member of the gastric microbiota in over half of the human population of which 5-15% develop gastritis or gastric malignancies. Immune responses to H. pylori are characterized by mixed T helper cell, cytotoxic T cell and NK cell responses. The presence of Tregs is essential for the control of gastritis and together with regulatory CX3CR1+ mononuclear phagocytes and immune-evasion strategies they enable life-long persistence of H. pylori. This H. pylori-induced regulatory environment might contribute to its cross-protective effect in inflammatory bowel disease and obesity. Here we review host-microbe interactions, the development of pro- and anti-inflammatory immune responses and how the latter contribute to H. pylori's role as beneficial member of the gut microbiota. Furthermore, we present the integration of existing and new data into a computational/mathematical model and its use for the investigation of immunological mechanisms underlying initiation, progression and outcomes of H. pylori infection.
Collapse
Affiliation(s)
- Barbara Kronsteiner
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| | | | - Monica Viladomiu
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| | | | - Vida Abedi
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| |
Collapse
|
16
|
Deen NS, Gong L, Naderer T, Devenish RJ, Kwok T. Analysis of the Relative Contribution of Phagocytosis, LC3-Associated Phagocytosis, and Canonical Autophagy During Helicobacter pylori Infection of Macrophages. Helicobacter 2015; 20:449-59. [PMID: 25864465 DOI: 10.1111/hel.12223] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Previous findings have suggested that Helicobacter pylori induces autophagic processes and subsequently takes refuge in autophagosomes, thereby contributing to persistent infection. Recently, a noncanonical form of autophagy, LC3 (microtubule-associated protein 1 light chain 3)-associated phagocytosis (LAP), has been shown to be required for efficient clearance of some intracellular bacteria. Whether H. pylori infection induces LAP had not been examined previously. In this study, we determined the extent to which H. pylori infection induces canonical autophagy or LAP in macrophages, and the involvement of the H. pylori cag pathogenicity island (cagPAI) with these processes. METHODS Immunofluorescence confocal microscopy was used to analyze the formation of GFP-LC3 puncta and their colocalization with H. pylori. Transmission electron microscopy was used to detect the ultrastructure of H. pylori-containing compartments. RESULTS The majority of intracellular bacteria (85-95%) were found in phagosomes that were LC3-negative, with a small proportion (4-14%) appearing "free" in the cytosol. Only a very small percentage (0.5-6%) of intracellular H. pylori was sequestered in autophagosomes. Furthermore, no statistically significant difference in the relative distribution of H. pylori in the various compartments was observed between wild-type and cagPAI-mutant bacteria. CONCLUSIONS In macrophages, H. pylori infection does not induce LAP, but can induce canonical autophagy, which entraps a very small fraction of intracellular bacteria. We propose that this subpopulation of intracellular H. pylori might have escaped from phagosomes into the cytosol before being sequestered by autophagosomes. The cagPAI of H. pylori has only minor influence, if any, on the extent of these processes.
Collapse
Affiliation(s)
- Nadia S Deen
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Vic., Australia
| | - Lan Gong
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Vic., Australia
| | - Thomas Naderer
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Vic., Australia
| | - Rodney J Devenish
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Vic., Australia
| | - Terry Kwok
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Vic., Australia.,Department of Microbiology, Monash University, Clayton, Vic., Australia
| |
Collapse
|
17
|
Naj X, Linder S. ER-Coordinated Activities of Rab22a and Rab5a Drive Phagosomal Compaction and Intracellular Processing of Borrelia burgdorferi by Macrophages. Cell Rep 2015; 12:1816-30. [DOI: 10.1016/j.celrep.2015.08.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 06/24/2015] [Accepted: 08/08/2015] [Indexed: 01/26/2023] Open
|
18
|
Devarajan PV, Jain S, Dutta R. Infectious Diseases: Need for Targeted Drug Delivery. TARGETED DRUG DELIVERY : CONCEPTS AND DESIGN 2014. [PMCID: PMC7122176 DOI: 10.1007/978-3-319-11355-5_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Infectious diseases are a leading cause of death worldwide, with the constant fear of global epidemics. It is indeed an irony that the reticuloendothelial system (RES), the body’s major defence system, is the primary site for intracellular infections which are more difficult to treat. Pro-inflammatory M1 macrophages play an important role in defence. However, ingenious pathogen survival mechanisms including phagolysosome destruction enable their persistence. Microbial biofilms present additional challenges. Low intracellular drug concentrations, drug efflux by efflux pumps and/or enzymatic degradation, emergence of multi-drug resistance (MDR), are serious limitations of conventional therapy. Targeted delivery using nanocarriers, and passive and active targeting strategies could provide quantum increase in intracellular drug concentration. Receptor mediated endocytosis using appropriate ligands is a viable approach. Liposomes and polymeric/lipidic nanoparticles, dendrimers micelles and micro/nanoemulsions could all be relied upon. Specialised targeting approaches are demonstrated for important diseases like tuberculosis, HIV and Malaria. Application of targeted delivery in the treatment of veterinary infections is exemplified and future possibilities indicated. The chapter thus provides an overview on important aspects of infectious diseases and the challenges therein, while stressing on the promise of targeted drug delivery in augmenting therapy of infectious diseases.
Collapse
Affiliation(s)
- Padma V. Devarajan
- grid.44871.3e0000000106680201Institute of Chemical Technology, Department of Pharmaceutical Sciences and Technology, Mumbai, India
| | - Sanyog Jain
- grid.419631.8000000008877852XNational Institute of Pharmaceutical Education and Research (NIPER), Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, Mohali, Punjab India
| | | |
Collapse
|
19
|
Ansari SA, Devi S, Tenguria S, Kumar A, Ahmed N. Helicobacter pylori protein HP0986 (TieA) interacts with mouse TNFR1 and triggers proinflammatory and proapoptotic signaling pathways in cultured macrophage cells (RAW 264.7). Cytokine 2014; 68:110-7. [DOI: 10.1016/j.cyto.2014.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 02/09/2014] [Accepted: 03/24/2014] [Indexed: 12/12/2022]
|
20
|
Du SY, Wang HJ, Cheng HH, Chen SD, Wang LHC, Wang WC. Cholesterol glucosylation by Helicobacter pylori delays internalization and arrests phagosome maturation in macrophages. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2014; 49:636-645. [PMID: 25070282 DOI: 10.1016/j.jmii.2014.05.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/16/2014] [Accepted: 05/29/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND/PURPOSE Helicobacter pylori colonizes the human stomach and contributes to chronic inflammation of the gastric mucosa. H. pylori persistence occurs because of insufficient eradication by phagocytic cells. A key factor of H. pylori, cholesterol-α-glucosyltransferase encoded by capJ that extracts host cholesterol and converts it to cholesteryl glucosides, is important to evade host immunity. Here, we examined whether phagocytic trafficking in macrophages was perturbed by capJ-carrying H. pylori. METHODS J774A.1 cells were infected with H. pylori at a multiplicity of infection of 50. Live-cell imaging and confocal microscopic analysis were applied to monitor the phagocytic trafficking events. The viability of H. pylori inside macrophages was determined by using gentamicin colony-forming unit assay. The phagocytic routes were characterized by using trafficking-intervention compounds. RESULTS Wild type (WT) H. pylori exhibited more delayed entry into macrophages and also arrested phagosome maturation more than did capJ knockout mutant. Pretreatment of genistein and LY294002 prior to H. pylori infection reduced the internalization of WT but not capJ-knockout H. pylori in macrophages. CONCLUSION Cholesterol glucosylation by H. pylori interferes with phagosome trafficking via a lipid-raft and PI3K-dependent manner, which retards engulfment of bacteria for prolonged intracellular survival of H. pylori.
Collapse
Affiliation(s)
- Shin-Yi Du
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Hung-Jung Wang
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsin-Hung Cheng
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan; Biomedical Science and Engineering Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Sheng-De Chen
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Lily Hui-Ching Wang
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Ching Wang
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan; Biomedical Science and Engineering Center, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
21
|
Kronsteiner B, Bassaganya-Riera J, Philipson N, Hontecillas R. Novel insights on the role of CD8+ T cells and cytotoxic responses during Helicobacter pylori infection. Gut Microbes 2014; 5:357-62. [PMID: 24755940 PMCID: PMC4153774 DOI: 10.4161/gmic.28899] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori chronically persists in 50% of the human population and causes serious gastric and duodenal pathologies in 15% of infected people. Research on the immune response to the infection has mainly focused on the induction of CD4+ T cell responses. Human studies emphasize the potential clinical relevance of CD8+ cytotoxic T lymphocytes, however this cell type has barely been reported in studies employing mouse or gerbil models. Traditionally characterized as an extracellular bacterium, H. pylori has been identified inside epithelial and immune cells. Similarly to other intracellular bacteria, H. pylori infection of macrophages can alter autophagy and phagosome processing. A novel animal model of H. pylori infection demonstrates for the first time the induction of cytotoxic CD8+ T cell responses in pigs and localization of intracellular H. pylori within lymphoid aggregates. Here, we discuss novel mechanisms of host-H. pylori interactions that could lead to the induction of cytotoxic responses.
Collapse
|
22
|
Helicobacter pylori infection in a pig model is dominated by Th1 and cytotoxic CD8+ T cell responses. Infect Immun 2013; 81:3803-13. [PMID: 23897614 DOI: 10.1128/iai.00660-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori infection is the leading cause for peptic ulcer disease and gastric adenocarcinoma. Mucosal T cell responses play an important role in mediating H. pylori-related gastric immunopathology. While induced regulatory T (iTreg) cells are required for chronic colonization without disease, T helper 1 (Th1) effector responses are associated with lower bacterial loads at the expense of gastric pathology. Pigs were inoculated with either H. pylori strain SS1 or J99. Phenotypic and functional changes in peripheral blood mononuclear cell (PBMC) populations were monitored weekly, and mucosal immune responses and bacterial loads were assessed up to 2 months postinfection. Both H. pylori strains elicited a Th1 response characterized by increased percentages of CD4(+)Tbet(+) cells and elevated gamma interferon (IFN-γ) mRNA in PBMCs. A subset of CD8(+) T cells expressing Tbet and CD16 increased following infection. Moreover, a significant increase in perforin and granzyme mRNA expression was observed in PBMCs of infected pigs, indicating a predominant cytotoxic immune response. Infiltration of B cells, myeloid cells, T cells expressing Treg- and Th17-associated transcription factors, and cytotoxic T cells was found in the gastric lamina propria of both infected groups. Interestingly, based on bacterial reisolation data, strain SS1 showed greater capacity to colonize and/or persist in the gastric mucosa than did strain J99. This novel pig model of infection closely mimics human gastric pathology and presents a suitable avenue for studying effector and regulatory responses toward H. pylori described in humans.
Collapse
|
23
|
Abstract
Phagocytosis and phagosome maturation are crucial processes in biology. Phagocytosis and the subsequent digestion of phagocytosed particles occur across a huge diversity of eukaryotes and can be achieved by many different cells within one organism. In parallel, diverse groups of pathogens have evolved mechanisms to avoid killing by phagocytic cells. The present review discusses a key innate immune cell, the macrophage, and highlights the myriad mechanisms microbes have established to escape phagocytic killing.
Collapse
Affiliation(s)
- Leanne M Smith
- Institute of Microbiology and Infection, School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | | |
Collapse
|
24
|
Borlace GN, Keep SJ, Prodoehl MJR, Jones HF, Butler RN, Brooks DA. A role for altered phagosome maturation in the long-term persistence of Helicobacter pylori infection. Am J Physiol Gastrointest Liver Physiol 2012; 303:G169-79. [PMID: 22575220 DOI: 10.1152/ajpgi.00320.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The vigorous host immune response that is mounted against Helicobacter pylori is unable to eliminate this pathogenic bacterium from its niche in the human gastric mucosa. This results in chronic inflammation, which can develop into gastric or duodenal ulcers in 10% of infected individuals and gastric cancer in 1% of infections. The determinants for these more severe pathologies include host (e.g., high IL-1β expression polymorphisms), bacterial [e.g., cytotoxicity-associated gene (cag) pathogenicity island], and environmental (e.g., dietary nitrites) factors. However, it is the failure of host immune effector cells to eliminate H. pylori that underlies its persistence and the subsequent H. pylori-associated disease. Here we discuss the mechanisms used by H. pylori to survive the host immune response and, in particular, the role played by altered phagosome maturation.
Collapse
Affiliation(s)
- Glenn N Borlace
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, Univ. of South Australia, South Australia 5001, Australia.
| | | | | | | | | | | |
Collapse
|
25
|
Yang ZX, Lu CY, Yang B, Xia N, Dou KF. PPK knockout attenuates evasion of immune elimination of Helicobacter pylori by macrophages. Shijie Huaren Xiaohua Zazhi 2012; 20:22-26. [DOI: 10.11569/wcjd.v20.i1.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the impact of knockout of the polyphosphate kinase gene in Helicobacter pylori (H. pylori) on bacterial evasion of immune elimination by macrophages.
METHODS: A PPK null mutant of H. pylori was constructed by gene homologous recombination. The polyphosphate was extracted from the PPK null mutant and wild type bacteria to compare the amount of polyphosphate by conversion into ATP. PPK null mutant H. pylori or wild type bacteria were co-cultured with murine macrophage cell line Raw 264.1 to compare the bacterial survival in macrophages at 24 h.
RESULTS: A PPK null mutant H. pylori strain was successfully constructed. The amount of polyphosphate in PPK null mutant bacteria was significantly lower than that in wild type bacteria (0.46 nmol Pi/mg Protein ± 0.25 nmol Pi/mg Protein vs 175.33 nmol Pi/mg Protein ± 21.22 nmol Pi/mg Protein, P < 0.01). Compared to wild type H. pylori, the survival rate of PPK null mutant bacteria in macrophages was similar at 2 h but was significantly reduced at 24 h.
CONCLUSION: PPK plays a critical role in synthesizing polyphosphate in H. pylori. PPK knockout in H. pylori significantly impaired their ability to synthesize polyphosphate and to evade immune elimination by macrophages.
Collapse
|