1
|
Barminga D, Mutinda S, Mobegi FM, Kibet W, Hale B, Anami S, Wijeratne A, Bellis ES, Runo S. Cell Wall Dynamics in the Parasitic Plant ( Striga) and Rice Pathosystem. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2025; 38:285-296. [PMID: 39636280 DOI: 10.1094/mpmi-06-24-0064-fi] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
In the plant-plant pathosystem of rice (Oryza sativa) and the parasitic plant Striga hermonthica, cell walls from either plant are important defensive and offensive structures. Here, we reveal the cell wall dynamics in both Striga and rice using simultaneous RNA sequencing. We used weighted gene co-expression network analysis to home in on cell wall modification processes occurring in interactions with a resistant rice cultivar (Nipponbare) compared with a susceptible one (IAC 165). Likewise, we compared the cell wall dynamics in Striga infecting resistant and susceptible rice. Our study revealed an intense battlement at the Striga-rice cell walls involving both parasite (offense) and host (defense) factors, the outcome of which makes the difference between successful or failed parasitism. Striga activates genes encoding cell wall-degrading enzymes to gain access to the host, expansins to allow for cell elongation, and pectin methyl esterase inhibitors for rigidity during infection. In the susceptible host, immune response processes are not induced, and Striga-derived cell wall-degrading enzymes easily breach the host cell wall, resulting in successful parasitism. In contrast, the resistant host invokes immune responses modulated by phytohormones to fortify the cell wall through polysaccharides and lignin deposition. Through these processes, the cell wall of the resistant host successfully obstructs parasite entry. We discuss the implications of these findings in the context of practical agriculture in which cell wall modification can be used to manage parasitic plants. [Formula: see text] Copyright © 2025 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.
Collapse
Affiliation(s)
- Damaris Barminga
- Institute of Biotechnology Research, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Sylvia Mutinda
- Pan African University Institute for Basic Sciences, Technology and Innovation, Nairobi, Kenya
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Fredrick M Mobegi
- Department of Clinical Immunology, PathWest Laboratory Medicine WA, Fiona Stanley Hospital Network, Murdoch, WA, Australia
- Department of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Willy Kibet
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Brett Hale
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR, U.S.A
| | - Sylvester Anami
- Institute of Biotechnology Research, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Asela Wijeratne
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR, U.S.A
| | - Emily S Bellis
- Department of Computer Science, Arkansas State University, Jonesboro, AR, U.S.A
| | - Steven Runo
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| |
Collapse
|
2
|
Li S, Hao L, Deng J, Zhang J, Yu F, Ye F, Li N, Hu X. The Culprit Behind HBV-Infected Hepatocytes: NTCP. Drug Des Devel Ther 2024; 18:4839-4858. [PMID: 39494152 PMCID: PMC11529284 DOI: 10.2147/dddt.s480151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Hepatitis B virus (HBV) is a globally prevalent human DNA virus responsible for over 250 million cases of chronic liver infections, leading to conditions such as liver inflammation, cirrhosis and hepatocellular carcinoma (HCC). Sodium taurocholate co-transporting polypeptide (NTCP) is a transmembrane protein highly expressed in human hepatocytes and functions as a bile acid (BA) transporter. NTCP has been identified as the receptor that HBV and its satellite virus, hepatitis delta virus (HDV), use to enter hepatocytes. HBV entry into hepatocytes is tightly regulated by various signaling pathways, and NTCP plays an important role as the initial stage of HBV infection. NTCP acts as an initiation signal, causing metabolic changes in hepatocytes and facilitating the entry of HBV into hepatocytes. Thus, a comprehensive understanding of NTCP's role is crucial. In this review, we will examine the regulatory mechanisms governing HBV pre-S1 binding to liver membrane NTCP, the role of NTCP in HBV internalization, and the transcriptional and translational regulation of NTCP expression. Additionally, we will discuss clinical drugs targeting NTCP, including combination therapies involving NTCP inhibitors, and consider the safety of NTCP as a therapeutic target.
Collapse
Affiliation(s)
- Shenghao Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Liyuan Hao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Jiali Deng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Junli Zhang
- Department of Infectious Diseases, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu Province, People’s Republic of China
| | - Fei Yu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Fanghang Ye
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Na Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
3
|
Junaid M, Lu H, Li Y, Liu Y, Din AU, Qi Z, Xiong Y, Yan J. Novel Synergistic Probiotic Intervention: Transcriptomic and Metabolomic Analysis Reveals Ameliorative Effects on Immunity, Gut Barrier, and Metabolism of Mice during Salmonella typhimurium Infection. Genes (Basel) 2024; 15:435. [PMID: 38674370 PMCID: PMC11050207 DOI: 10.3390/genes15040435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Salmonella typhimurium (S. typhimurium), a prevalent cause of foodborne infection, induces significant changes in the host transcriptome and metabolome. The lack of therapeutics with minimal or no side effects prompts the scientific community to explore alternative therapies. This study investigates the therapeutic potential of a probiotic mixture comprising Lactobacillus acidophilus (L. acidophilus 1.3251) and Lactobacillus plantarum (L. plantarum 9513) against S. typhimurium, utilizing transcriptome and metabolomic analyses, a novel approach that has not been previously documented. Twenty-four SPF-BALB/c mice were divided into four groups: control negative group (CNG); positive control group (CPG); probiotic-supplemented non-challenged group (LAPG); and probiotic-supplemented Salmonella-challenged group (LAPST). An RNA-sequencing analysis of small intestinal (ileum) tissue revealed 2907 upregulated and 394 downregulated DEGs in the LAPST vs. CPG group. A functional analysis of DEGs highlighted their significantly altered gene ontology (GO) terms related to metabolism, gut integrity, cellular development, and immunity (p ≤ 0.05). The KEGG analysis showed that differentially expressed genes (DEGs) in the LAPST group were primarily involved in pathways related to gut integrity, immunity, and metabolism, such as MAPK, PI3K-Akt, AMPK, the tryptophan metabolism, the glycine, serine, and threonine metabolism, ECM-receptor interaction, and others. Additionally, the fecal metabolic analysis identified 1215 upregulated and 305 downregulated metabolites in the LAPST vs. CPG group, implying their involvement in KEGG pathways including bile secretion, propanoate metabolism, arginine and proline metabolism, amino acid biosynthesis, and protein digestion and absorption, which are vital for maintaining barrier integrity, immunity, and metabolism. In conclusion, these findings suggest that the administration of a probiotic mixture improves immunity, maintains gut homeostasis and barrier integrity, and enhances metabolism in Salmonella infection.
Collapse
Affiliation(s)
- Muhammad Junaid
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Hongyu Lu
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Yixiang Li
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Yu Liu
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Ahmad Ud Din
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Yi Xiong
- Guangxi Center for Animals Disease Control and Prevention, Nanning 530004, China
| | - Jianhua Yan
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| |
Collapse
|
4
|
Tang H, Zhang D, Jiang F, Yu L, Tang H, Zhu J, Wu S, Niu H. Enhancement of Cell Adhesion by Anaplasma phagocytophilum Nucleolin-Interacting Protein AFAP. J Pers Med 2023; 13:jpm13020302. [PMID: 36836536 PMCID: PMC9965380 DOI: 10.3390/jpm13020302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Anaplasma phagocytophilum, the aetiologic agent of human granulocytic anaplasmosis (HGA), is an obligate intracellular Gram-negative bacterium. During infection, A. phagocytophilum enhances the adhesion of neutrophils to the infected endothelial cells. However, the bacterial factors contributing to this phenomenon remain unknown. In this study, we characterized a type IV secretion system substrate of A. phagocytophilum, AFAP (an actin filament-associated Anaplasma phagocytophilum protein) and found that it dynamically changed its pattern and subcellular location in cells and enhanced cell adhesion. Tandem affinity purification combined with mass spectrometry identified host nucleolin as an AFAP-interacting protein. Further study showed the disruption of nucleolin by RNA interference, and the treatment of a nucleolin-binding DNA aptamer AS1411 attenuated AFAP-mediated cell adhesion, indicating that AFAP enhanced cell adhesion in a nucleolin-dependent manner. The characterization of cell adhesion-enhancing AFAP and the identification of host nucleolin as its interaction partner may help understand the mechanism underlying A. phagocytophilum-promoting cell adhesion, facilitating the elucidation of HGA pathogenesis.
Collapse
Affiliation(s)
- Hongcheng Tang
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Daxiu Zhang
- Clinical Laboratory Center, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Fenfen Jiang
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Lifeng Yu
- Clinical Laboratory Center, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Hui Tang
- Clinical Laboratory Center, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Jiafeng Zhu
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Shuyan Wu
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Hua Niu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin 541001, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
- Correspondence:
| |
Collapse
|
5
|
Persistence Infection of TGEV Promotes Enterococcus faecalis Infection on IPEC-J2 Cells. Int J Mol Sci 2022; 24:ijms24010450. [PMID: 36613893 PMCID: PMC9820250 DOI: 10.3390/ijms24010450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
Transmissible gastroenteritis virus (TGEV) is a coronavirus causing diarrhea with high incidence in swine herds. Its persistent infection might lead to epithelial-mesenchymal transition (EMT) of swine intestinal epithelial cells, followed by subsequent infections of other pathogens. Enterococcus faecalis (E. faecalis) is a member of the enteric microorganisms and an opportunistic pathogen. There is no report of secondary E. faecalis infection to TGEV, even though they both target to the intestinal tracts. To investigate the interactions between TGEV and E. faecalis, we set up an in vitro infection model by the swine IPEC-J2 cells. Dynamic changes of cell traits, including EMT and cell motility, were evaluated through qPCR, Western blot, electronic microscopy, scratch test, Transwell migration test and invasion test, respectively. The adhesion and invasion tests of E. faecalis were taken to verify the impact of the preceding TGEV infection. The cell morphology and molecular marker evaluation results showed that the TGEV persistent infection induced EMT on IPEC-J2 cells; increased cellular motility and invasion potential were also observed. Spontaneously, the expression levels of fibronectin (FN) and the membrane protein integrin-α5, which are dominant bacterial receptors on IPEC-J2 cells, were upgraded. It indicated that the bacteria E. faecalis adhered to IPEC-J2 cells through the FN receptor, and then invaded the cells by binding with the integrin-α5, suggesting that both molecules were critical for the adhesion and invasion of E. faecalis to IPEC-J2 cells. Additionally, it appeared that E. faecalis alone might trigger certain EMT phenomena, implying a vicious circle might occur. Generally, bacterial and viral co-infections are frustrating yet common in both human and veterinary medicines, and our observations on enteric TGEV and E. faecalis interactions, especially the diversity of bacterial invasion strategies, might provide new insights into the mechanisms of E. faecalis pathogenicity.
Collapse
|
6
|
Kavuluko J, Kibe M, Sugut I, Kibet W, Masanga J, Mutinda S, Wamalwa M, Magomere T, Odeny D, Runo S. GWAS provides biological insights into mechanisms of the parasitic plant (Striga) resistance in sorghum. BMC PLANT BIOLOGY 2021; 21:392. [PMID: 34418971 PMCID: PMC8379865 DOI: 10.1186/s12870-021-03155-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/02/2021] [Indexed: 05/05/2023]
Abstract
BACKGROUND Sorghum yields in sub-Saharan Africa (SSA) are greatly reduced by parasitic plants of the genus Striga (witchweed). Vast global sorghum genetic diversity collections, as well as the availability of modern sequencing technologies, can be potentially harnessed to effectively manage the parasite. RESULTS We used laboratory assays - rhizotrons to screen a global sorghum diversity panel to identify new sources of resistance to Striga; determine mechanisms of resistance, and elucidate genetic loci underlying the resistance using genome-wide association studies (GWAS). New Striga resistant sorghum determined by the number, size and biomass of parasite attachments were identified. Resistance was by; i) mechanical barriers that blocked parasite entry, ii) elicitation of a hypersensitive reaction that interfered with parasite development, and iii) the inability of the parasite to develop vascular connections with hosts. Resistance genes underpinning the resistance corresponded with the resistance mechanisms and included pleiotropic drug resistance proteins that transport resistance molecules; xylanase inhibitors involved in cell wall fortification and hormonal regulators of resistance response, Ethylene Response Factors. CONCLUSIONS Our findings are of fundamental importance to developing durable and broad-spectrum resistance against Striga and have far-reaching applications in many SSA countries where Striga threatens the livelihoods of millions of smallholder farmers that rely on sorghum as a food staple.
Collapse
Affiliation(s)
- Jacinta Kavuluko
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Magdaline Kibe
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Irine Sugut
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Willy Kibet
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Joel Masanga
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Sylvia Mutinda
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
- Pan African University of Science Technology and Innovation, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Mark Wamalwa
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Titus Magomere
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Damaris Odeny
- International Crops Research Institute for the Semi-Arid Tropics, Nairobi, Kenya
| | - Steven Runo
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya.
| |
Collapse
|
7
|
Memon FU, Yang Y, Leghari IH, Lv F, Soliman AM, Zhang W, Si H. Transcriptome Analysis Revealed Ameliorative Effects of Bacillus Based Probiotic on Immunity, Gut Barrier System, and Metabolism of Chicken under an Experimentally Induced Eimeria tenella Infection. Genes (Basel) 2021; 12:genes12040536. [PMID: 33917156 PMCID: PMC8067821 DOI: 10.3390/genes12040536] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/03/2021] [Accepted: 04/04/2021] [Indexed: 12/20/2022] Open
Abstract
In this study, we performed transcriptome analysis in the cecum tissues of negative control untreated non-challenged (NC), positive control untreated challenged (PC), and Bacillus subtilis (B. subtilis) fed challenged chickens (BS + ET) in order to examine the underlying potential therapeutic mechanisms of Bacillus based probiotic feeding under an experimental Eimeria tenella (E. tenella) infection. Our results for clinical parameters showed that birds in probiotic diet decreased the bloody diarrhea scores, oocyst shedding, and lesion scores compared to positive control birds. RNA-sequencing (RNA-seq) analysis revealed that in total, 2509 up-regulated and 2465 down-regulated differentially expressed genes (DEGs) were detected in the PC group versus NC group comparison. In the comparison of BS + ET group versus PC group, a total of 784 up-regulated and 493 down-regulated DEGs were found. Among them, several DEGs encoding proteins involved in immunity, gut barrier integrity, homeostasis, and metabolism were up-regulated by the treatment of probiotic. Functional analysis of DEGs also revealed that some gene ontology (GO) terms related with immunity, metabolism and cellular development were significantly affected by the exposure of probiotic. Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis showed that the DEGs in the cecum of B. subtilis-fed challenged group were mainly participated in the pathways related with immunity and gut barrier integrity, included mitogen-activated protein kinase (MAPK) signaling pathway, toll-like receptor (TLR) signaling pathway, extracellular matrix (ECM)–receptor interaction, tight junction, and so on. Taken together, these results suggest that Bacillus based probiotic modulate the immunity, maintain gut homeostasis as well as barrier system and improve chicken metabolism during E. tenella infection.
Collapse
Affiliation(s)
- Fareed Uddin Memon
- College of Animal Sciences and Technology, Guangxi University, Nanning 530004, China; (F.U.M.); (Y.Y.); (F.L.); (A.M.S.); (W.Z.)
- Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Tando Jam 70060, Pakistan;
| | - Yunqiao Yang
- College of Animal Sciences and Technology, Guangxi University, Nanning 530004, China; (F.U.M.); (Y.Y.); (F.L.); (A.M.S.); (W.Z.)
| | - Imdad Hussain Leghari
- Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Tando Jam 70060, Pakistan;
| | - Feifei Lv
- College of Animal Sciences and Technology, Guangxi University, Nanning 530004, China; (F.U.M.); (Y.Y.); (F.L.); (A.M.S.); (W.Z.)
| | - Ahmed M. Soliman
- College of Animal Sciences and Technology, Guangxi University, Nanning 530004, China; (F.U.M.); (Y.Y.); (F.L.); (A.M.S.); (W.Z.)
- Agricultural Research Center, Biotechnology Department, Animal Health Research Institute, Giza 12618, Egypt
| | - Weiyu Zhang
- College of Animal Sciences and Technology, Guangxi University, Nanning 530004, China; (F.U.M.); (Y.Y.); (F.L.); (A.M.S.); (W.Z.)
| | - Hongbin Si
- College of Animal Sciences and Technology, Guangxi University, Nanning 530004, China; (F.U.M.); (Y.Y.); (F.L.); (A.M.S.); (W.Z.)
- Correspondence:
| |
Collapse
|
8
|
Li C, Bai X, Liu X, Zhang Y, Liu L, Zhang L, Xu F, Yang Y, Liu M. Disruption of Epithelial Barrier of Caco-2 Cell Monolayers by Excretory Secretory Products of Trichinella spiralis Might Be Related to Serine Protease. Front Microbiol 2021; 12:634185. [PMID: 33815318 PMCID: PMC8013981 DOI: 10.3389/fmicb.2021.634185] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/19/2021] [Indexed: 11/16/2022] Open
Abstract
The physical barrier is composed of epithelial cells which are joined together through intercellular connections. It serves to prevent pathogenic microorganisms from departing the intestinal lumen to invade the host. The excretory secretory (ES) products of Trichinella spiralis are critical for invasion. However, whether ES products of T. spiralis can act on the intestinal barrier is still unknown. In this study, the role of ES products of T. spiralis muscle larvae (Ts-ML-ES) in host invasion was studied by establishing an in vitro cell monolayers model. Barrier integrity analysis by a transmembrane resistance test and a paracellular permeability assay revealed that the Ts-ML-ES was able to destroy barrier function. It occurred via a reduction in the expression of tight junction (TJ) proteins, which was induced by serine protease. Furthermore, Western bolt analysis indicated that Ts-ML-ES reduced the expression of TJ proteins via the MAPK signaling pathway. Based on these data, we conclude that serine protease are likely the main factors from Ts-ML-ES that affect host intestinal barrier integrity by reducing the expression of TJs via the P38-MAPK signaling pathway. Serine protease in Ts-ML-ES might be a key invasion factor in T. spiralis.
Collapse
Affiliation(s)
- Chengyao Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xue Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yuanyuan Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Lei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Lixiao Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Fengyan Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yong Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
9
|
Lima PC, Hartley-Tassell L, Cooper O, Wynne JW. Searching for the sweet spot of amoebic gill disease of farmed Atlantic salmon: the potential role of glycan-lectin interactions in the adhesion of Neoparamoeba perurans. Int J Parasitol 2021; 51:545-557. [PMID: 33675796 DOI: 10.1016/j.ijpara.2020.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 01/25/2023]
Abstract
One of the first critical steps in the pathogenesis of amoebic gill disease (AGD) of farmed salmon is the adhesion of the causative amoeba to the host. The current study aimed to investigate the potential involvement of glycan-binding proteins expressed on the extracellular surface of Neoparamoeba perurans in gill tissue recognition and binding. The glycan-binding properties of the surface membrane of N. perurans and the carbohydrate binding profile of Atlantic salmon gill-derived epithelial cells were identified through the use of glycan and lectin microarrays, respectively. The occurrence of specific carbohydrate-mediated binding was then further assessed by in vitro attachment assays using microtitre plates pre-coated with the main glycan candidates. Adhesion assays were also performed in the presence of exogenous saccharides with the aim of blocking glycan-specific binding activity. Comparative analysis of the results from both lectin and glycan arrays showed significant overlap, as some glycans to which binding by the amoeba was seen were reflected as being present on the gill epithelial cells. The two main candidates proposed to be involved in amoeba attachment to the gills are mannobiose and N-acetylgalactosamine (GalNAc). Adhesion of amoebae significantly increased by 33.5 and 23% when cells were added to α1,3-Mannobiose-BSA and GalNAc-BSA coated plates. The observed increased in attachment was significantly reduced when the amoebae were incubated with exogenous glycans, further demonstrating the presence of mannobiose- and GalNAc-binding sites on the surfaces of the cells. We believe this study provides the first evidence for the presence of a highly specific carbohydrate recognition and binding system in N. perurans. These preliminary findings could be of extreme importance given that AGD is an external parasitic infestation and much of the current research on the development of alternative treatment strategies relies on either instant amoeba detachment or blocking parasite attachment.
Collapse
Affiliation(s)
- P C Lima
- CSIRO Agriculture and Food, Livestock & Aquaculture, Queensland Biosciences Precinct, 306 Carmody Road, Brisbane, QLD 4067, Australia.
| | - L Hartley-Tassell
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, QLD 4222, Australia
| | - O Cooper
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, QLD 4222, Australia
| | - J W Wynne
- CSIRO Agriculture and Food, Livestock & Aquaculture, Castray Esplanade, Battery Point, TAS 7004, Australia
| |
Collapse
|
10
|
Cheudjeu A. Correlation of D-xylose with severity and morbidity-related factors of COVID-19 and possible therapeutic use of D-xylose and antibiotics for COVID-19. Life Sci 2020; 260:118335. [PMID: 32846167 PMCID: PMC7443215 DOI: 10.1016/j.lfs.2020.118335] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 01/08/2023]
Abstract
The SARS-Cov-2 pandemic that currently affects the entire world has been shown to be especially dangerous in the elderly (≥65 years) and in smokers, with notably strong comorbidity in patients already suffering from chronic diseases, such as Type 2 diabetes, cancers, chronic respiratory diseases, obesity, and hypertension. Inflammation of the lungs is the main factor leading to respiratory distress in patients with chronic respiratory disease and in patients with severe COVID-19. Several studies have shown that inflammation of the lungs in general and Type 2 diabetes are accompanied by the degradation of glycosaminoglycans (GAGs), especially heparan sulfate (HS). Several studies have also shown the importance of countering the degradation of HS in lung infections and Type 2 diabetes. D-xylose, which is the initiating element for different sulfate GAG chains (especially HS), has shown regeneration properties for GAGs. D-xylose and xylitol have demonstrated anti-inflammatory, antiglycemic, antiviral, and antibacterial properties in lung infections, alone or in combination with antibiotics. Considering the existing research on COVID-19 and related to D-xylose/xylitol, this review offers a perspective on why the association between D-xylose and antibiotics may contribute to significantly reducing the duration of treatment of COVID-19 patients and why some anti-inflammatory drugs may increase the severity of COVID-19. A strong correlation with scurvy, based on gender, age, ethnicity, smoking status, and obesity status, is also reviewed. Related to this, the effects of treatment with plants such as Artemisia are also addressed. CHEMICAL COMPOUNDS: D-xylose; xylitol; l-ascorbic Acid; D-glucuronic acid; N-acetylglucosamine; D-N-acetylglucosamine; N-acetylgalactosamine; galactose.
Collapse
|
11
|
Hu Q, Zhang F, Duan L, Wang B, Ye Y, Li P, Li D, Yang S, Zhou L, Chen W. E-cadherin Plays a Role in Hepatitis B Virus Entry Through Affecting Glycosylated Sodium-Taurocholate Cotransporting Polypeptide Distribution. Front Cell Infect Microbiol 2020; 10:74. [PMID: 32175289 PMCID: PMC7056903 DOI: 10.3389/fcimb.2020.00074] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a major cause of chronic liver disease and hepatocellular carcinoma. Current antiviral therapy does not effectively eradicate HBV and further investigations into the mechanisms of viral infection are needed to enable the development of new therapeutic agents. The sodium-taurocholate cotransporting polypeptide (NTCP) has been identified as a functional receptor for HBV entry in liver cells. However, the NTCP receptor is not sufficient for entry and other membrane proteins contribute to modulate HBV entry. This study seeks to understand how the NTCP functions in HBV entry. Herein we show that knockdown of the cell-cell adhesion molecule, E-cadherin significantly reduced infection by HBV particles and entry by HBV pseudoparticles in infected liver cells and cell lines. The glycosylated NTCP localizes to the plasma membrane through interaction with E- cadherin, which increases interaction with the preS1 portion of the Large HBV surface antigen. Our study contributes novel insights that advance knowledge of HBV infection at the level of host cell binding and viral entry.
Collapse
Affiliation(s)
- Qin Hu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics of Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Feifei Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liang Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanyuan Ye
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Pu Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dandan Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengjun Yang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Zhou
- Key Laboratory of Laboratory Medical Diagnostics of Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Weixian Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Dolat L, Valdivia RH. A renewed tool kit to explore Chlamydia pathogenesis: from molecular genetics to new infection models. F1000Res 2019; 8. [PMID: 31249676 PMCID: PMC6589931 DOI: 10.12688/f1000research.18832.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2019] [Indexed: 12/21/2022] Open
Abstract
Chlamydia trachomatis is the most prevalent sexually transmitted bacterial pathogen and the leading cause of preventable blindness in the developing world.
C. trachomatis invades the epithelium of the conjunctiva and genital tract and replicates within an intracellular membrane-bound compartment termed the inclusion. To invade and replicate in mammalian cells,
Chlamydia remodels epithelial surfaces by reorganizing the cytoskeleton and cell–cell adhesions, reprograms membrane trafficking, and modulates cell signaling to dampen innate immune responses. If the infection ascends to the upper female genital tract, it can result in pelvic inflammatory disease and tissue scarring.
C. trachomatis infections are associated with infertility, ectopic pregnancies, the fibrotic disorder endometriosis, and potentially cancers of the cervix and uterus. Unfortunately, the molecular mechanisms by which this clinically important human pathogen subverts host cellular functions and causes disease have remained relatively poorly understood because of the dearth of molecular genetic tools to study
Chlamydiae and limitations of both
in vivo and
in vitro infection models. In this review, we discuss recent advances in the experimental molecular tool kit available to dissect
C. trachomatis infections with a special focus on
Chlamydia-induced epithelial barrier disruption by regulating the structure, function, and dynamics of epithelial cell–cell junctions.
Collapse
Affiliation(s)
- Lee Dolat
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, USA
| | - Raphael H Valdivia
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, USA
| |
Collapse
|
13
|
Elumalai P, Rubeena AS, Arockiaraj J, Wongpanya R, Cammarata M, Ringø E, Vaseeharan B. The Role of Lectins in Finfish: A Review. REVIEWS IN FISHERIES SCIENCE & AQUACULTURE 2019; 27:152-169. [DOI: 10.1080/23308249.2018.1520191] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2023]
Affiliation(s)
- Preetham Elumalai
- School of Processing Technology, Kerala University of Fisheries and Ocean Studies, Panangad, Kerala, India
| | - Abdul Salam Rubeena
- School of Ocean Science and Technology, Kerala University of Fisheries and Ocean Studies, Panangad, Kerala, India
| | - Jesu Arockiaraj
- SRM Research Institute, SRM Institute of Science and Technology (Formerly known as SRM University), Kattankulathur, Chennai, Tamil Nadu, India
| | - Ratree Wongpanya
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Matteo Cammarata
- Marine Immunobiology Laboratory, Department of Earth and Marine Science, University of Palermo, Palermo, Italy
| | - Einar Ringø
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Baskaralingam Vaseeharan
- Crustacean Molecular Biology and Genomics Division, Biomaterials and Biotechnology in Animal Health Lab, Department of Animal Health and Management, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
14
|
Yeh SJ, Yeh CC, Lan CY, Chen BS. Investigating Common Pathogenic Mechanisms between Homo sapiens and Different Strains of Candida albicans for Drug Design: Systems Biology Approach via Two-Sided NGS Data Identification. Toxins (Basel) 2019; 11:toxins11020119. [PMID: 30769958 PMCID: PMC6409619 DOI: 10.3390/toxins11020119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 01/15/2023] Open
Abstract
Candida albicans (C. albicans) is the most prevalent fungal species. Although it is a healthy microbiota, genetic and epigenetic alterations in host and pathogen, and microenvironment changes would lead to thrush, vaginal yeast infection, and even hematogenously disseminated infection. Despite the fact that cytotoxicity is well-characterized, few studies discuss the genome-wide genetic and epigenetic molecular mechanisms between host and C. albicans. The aim of this study is to identify drug targets and design a multiple-molecule drug to prevent the infection from C. albicans. To investigate the common and specific pathogenic mechanisms in human oral epithelial OKF6/TERT-2 cells during the C. albicans infection in different strains, systems modeling and big databases mining were used to construct candidate host–pathogen genetic and epigenetic interspecies network (GEIN). System identification and system order detection are applied on two-sided next generation sequencing (NGS) data to build real host–pathogen cross-talk GEINs. Core host–pathogen cross-talk networks (HPCNs) are extracted by principal network projection (PNP) method. By comparing with core HPCNs in different strains of C. albicans, common pathogenic mechanisms were investigated and several drug targets were suggested as follows: orf19.5034 (YBP1) with the ability of anti-ROS; orf19.939 (NAM7), orf19.2087 (SAS2), orf19.1093 (FLO8) and orf19.1854 (HHF22) with high correlation to the hyphae growth and pathogen protein interaction; orf19.5585 (SAP5), orf19.5542 (SAP6) and orf19.4519 (SUV3) with the cause of biofilm formation. Eventually, five corresponding compounds—Tunicamycin, Terbinafine, Cerulenin, Tetracycline and Tetrandrine—with three known drugs could be considered as a potential multiple-molecule drug for therapeutic treatment of C. albicans.
Collapse
Affiliation(s)
- Shan-Ju Yeh
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Chun-Chieh Yeh
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Chung-Yu Lan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan.
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Bor-Sen Chen
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
- Department of Electrical Engineering, Yuan Ze University, Chungli 32003, Taiwan.
| |
Collapse
|
15
|
Wan L, Lin J, Du H, Zhang Y, Bravo A, Soberón M, Sun M, Peng D. Bacillus thuringiensistargets the host intestinal epithelial junctions for successful infection ofCaenorhabditis elegans. Environ Microbiol 2019; 21:1086-1098. [DOI: 10.1111/1462-2920.14528] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/02/2018] [Accepted: 12/12/2018] [Indexed: 01/28/2023]
Affiliation(s)
- Liting Wan
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural University Wuhan 430070 Hubei People's Republic of China
| | - Jian Lin
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural University Wuhan 430070 Hubei People's Republic of China
| | - Hongwen Du
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural University Wuhan 430070 Hubei People's Republic of China
| | - Yulan Zhang
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural University Wuhan 430070 Hubei People's Republic of China
| | - Alejandra Bravo
- Instituto de BiotecnologíaUniversidad Nacional Autónoma de México Apdo. postal 510‐3, Cuernavaca, 62250 Morelos Mexico
| | - Mario Soberón
- Instituto de BiotecnologíaUniversidad Nacional Autónoma de México Apdo. postal 510‐3, Cuernavaca, 62250 Morelos Mexico
| | - Ming Sun
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural University Wuhan 430070 Hubei People's Republic of China
| | - Donghai Peng
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural University Wuhan 430070 Hubei People's Republic of China
| |
Collapse
|
16
|
Modeling Host-Pathogen Interactions in the Context of the Microenvironment: Three-Dimensional Cell Culture Comes of Age. Infect Immun 2018; 86:IAI.00282-18. [PMID: 30181350 DOI: 10.1128/iai.00282-18] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tissues and organs provide the structural and biochemical landscapes upon which microbial pathogens and commensals function to regulate health and disease. While flat two-dimensional (2-D) monolayers composed of a single cell type have provided important insight into understanding host-pathogen interactions and infectious disease mechanisms, these reductionist models lack many essential features present in the native host microenvironment that are known to regulate infection, including three-dimensional (3-D) architecture, multicellular complexity, commensal microbiota, gas exchange and nutrient gradients, and physiologically relevant biomechanical forces (e.g., fluid shear, stretch, compression). A major challenge in tissue engineering for infectious disease research is recreating this dynamic 3-D microenvironment (biological, chemical, and physical/mechanical) to more accurately model the initiation and progression of host-pathogen interactions in the laboratory. Here we review selected 3-D models of human intestinal mucosa, which represent a major portal of entry for infectious pathogens and an important niche for commensal microbiota. We highlight seminal studies that have used these models to interrogate host-pathogen interactions and infectious disease mechanisms, and we present this literature in the appropriate historical context. Models discussed include 3-D organotypic cultures engineered in the rotating wall vessel (RWV) bioreactor, extracellular matrix (ECM)-embedded/organoid models, and organ-on-a-chip (OAC) models. Collectively, these technologies provide a more physiologically relevant and predictive framework for investigating infectious disease mechanisms and antimicrobial therapies at the intersection of the host, microbe, and their local microenvironments.
Collapse
|
17
|
Antunes S, Couto J, Ferrolho J, Rodrigues F, Nobre J, Santos AS, Santos-Silva MM, de la Fuente J, Domingos A. Rhipicephalus bursa Sialotranscriptomic Response to Blood Feeding and Babesia ovis Infection: Identification of Candidate Protective Antigens. Front Cell Infect Microbiol 2018; 8:116. [PMID: 29780749 PMCID: PMC5945973 DOI: 10.3389/fcimb.2018.00116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/23/2018] [Indexed: 02/03/2023] Open
Abstract
Ticks are among the most prevalent blood-feeding arthropods, and they act as vectors and reservoirs for numerous pathogens. Sialotranscriptomic characterizations of tick responses to blood feeding and pathogen infections can offer new insights into the molecular interplay occurring at the tick-host-pathogen interface. In the present study, we aimed to identify and characterize Rhipicephalus bursa salivary gland (SG) genes that were differentially expressed in response to blood feeding and Babesia ovis infection. Our experimental approach consisted of RNA sequencing of SG from three different tick samples, fed-infected, fed-uninfected, and unfed-uninfected, for characterization and inter-comparison. Overall, 7,272 expressed sequence tags (ESTs) were constructed from unfed-uninfected, 13,819 ESTs from fed-uninfected, and 15,292 ESTs from fed-infected ticks. Two catalogs of transcripts that were differentially expressed in response to blood feeding and B. ovis infection were produced. Four genes coding for a putative vitellogenin-3, lachesin, a glycine rich protein, and a secreted cement protein were selected for RNA interference functional studies. A reduction of 92, 65, and 51% was observed in vitellogenin-3, secreted cement, and lachesin mRNA levels in SG, respectively. The vitellogenin-3 knockdown led to increased tick mortality, with 77% of ticks dying post-infestation. The reduction of the secreted cement protein-mRNA levels resulted in 46% of ticks being incapable of correctly attaching to the host and significantly lower female weights post-feeding in comparison to the control group. The lachesin knockdown resulted in a 70% reduction of the levels associated with B. ovis infection in R. bursa SG and 70% mortality. These results improved our understanding of the role of tick SG genes in Babesia infection/proliferation and tick feeding. Moreover, lachesin, vitellogenin-3, and secreted cement proteins were validated as candidate protective antigens for the development of novel tick and tick-borne disease control measures.
Collapse
Affiliation(s)
- Sandra Antunes
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal.,Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Joana Couto
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal.,Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Joana Ferrolho
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal.,Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Fábio Rodrigues
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal
| | - João Nobre
- Instituto Nacional de Investigação Agrária e Veterinária, Pólo de Santarém, Vale de Santarém, Portugal
| | - Ana S Santos
- Instituto Nacional de Saúde Doutor Ricardo Jorge, Centro de Estudos de Vectores e Doenças Infecciosas Dr. Francisco Cambournac (CEVDI/INSA), Águas de Moura, Portugal
| | - M Margarida Santos-Silva
- Instituto Nacional de Saúde Doutor Ricardo Jorge, Centro de Estudos de Vectores e Doenças Infecciosas Dr. Francisco Cambournac (CEVDI/INSA), Águas de Moura, Portugal
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ciudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Ana Domingos
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal.,Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
18
|
Prashar A, Ortiz ME, Lucarelli S, Barker E, Tabatabeiyazdi Z, Shamoun F, Raju D, Antonescu C, Guyard C, Terebiznik MR. Small Rho GTPases and the Effector VipA Mediate the Invasion of Epithelial Cells by Filamentous Legionella pneumophila. Front Cell Infect Microbiol 2018; 8:133. [PMID: 29774203 PMCID: PMC5943596 DOI: 10.3389/fcimb.2018.00133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 04/17/2018] [Indexed: 12/13/2022] Open
Abstract
Legionella pneumophila (Lp) exhibits different morphologies with varying degrees of virulence. Despite their detection in environmental sources of outbreaks and in respiratory tract secretions and lung autopsies from patients, the filamentous morphotype of Lp remains poorly studied. We previously demonstrated that filamentous Lp invades lung epithelial cells (LECs) and replicates intracellularly in a Legionella containing vacuole. Filamentous Lp activates β1integrin and E-cadherin receptors at the surface of LECs leading to the formation of actin-rich cell membrane structures we termed hooks and membrane wraps. These structures entrap segments of an Lp filament on host cell surface and mediate bacterial internalization. Here we investigated the molecular mechanisms responsible for the actin rearrangements needed for the formation and elongation of these membrane wraps and bacterial internalization. We combined genetic and pharmacological approaches to assess the contribution of signaling downstream of β1integrin and E-cadherin receptors, and Lp Dot/Icm secretion system- translocated effectors toward the invasion process. Our studies demonstrate a multi-stage mechanism of LEC invasion by filamentous Lp. Bacterial attachment to host cells depends on signaling downstream of β1integrin and E-cadherin activation, leading to Rho GTPases-dependent activation of cellular actin nucleating proteins, Arp2/3 and mDia. This mediates the formation of primordial membrane wraps that entrap the filamentous bacteria on the cell surface. Following this, in a second phase of the invasion process the Dot/Icm translocated effector VipA mediates rapid membrane wrap elongation, leading to the engulfment of the filamentous bacteria by the LECs. Our findings provide the first description of Rho GTPases and a Dot/Icm effector VipA regulating the actin dynamics needed for the invasion of epithelial cells by Lp.
Collapse
Affiliation(s)
- Akriti Prashar
- Department of Biological Sciences, University of Toronto at Scarborough, Scarborough, ON, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - María Eugenia Ortiz
- Department of Biological Sciences, University of Toronto at Scarborough, Scarborough, ON, Canada
| | - Stefanie Lucarelli
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Elizabeth Barker
- Department of Biological Sciences, University of Toronto at Scarborough, Scarborough, ON, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Zohreh Tabatabeiyazdi
- Department of Biological Sciences, University of Toronto at Scarborough, Scarborough, ON, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Feras Shamoun
- Department of Biological Sciences, University of Toronto at Scarborough, Scarborough, ON, Canada
| | - Deepa Raju
- Department of Biological Sciences, University of Toronto at Scarborough, Scarborough, ON, Canada
| | - Costin Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Cyril Guyard
- Bioaster, Lyon, France.,Molecular Microbiology, Public Health Ontario, Toronto, ON, Canada
| | - Mauricio R Terebiznik
- Department of Biological Sciences, University of Toronto at Scarborough, Scarborough, ON, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
I love you Toxoplasma gondii. Microbes Infect 2017; 20:131-134. [PMID: 29248636 DOI: 10.1016/j.micinf.2017.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 11/20/2022]
|
20
|
Calderón-Garcidueñas L, Reynoso-Robles R, Pérez-Guillé B, Mukherjee PS, Gónzalez-Maciel A. Combustion-derived nanoparticles, the neuroenteric system, cervical vagus, hyperphosphorylated alpha synuclein and tau in young Mexico City residents. ENVIRONMENTAL RESEARCH 2017; 159:186-201. [PMID: 28803148 DOI: 10.1016/j.envres.2017.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/02/2017] [Accepted: 08/04/2017] [Indexed: 06/07/2023]
Abstract
Mexico City (MC) young residents are exposed to high levels of fine particulate matter (PM2.5), have high frontal concentrations of combustion-derived nanoparticles (CDNPs), accumulation of hyperphosphorylated aggregated α-synuclein (α-Syn) and early Parkinson's disease (PD). Swallowed CDNPs have easy access to epithelium and submucosa, damaging gastrointestinal (GI) barrier integrity and accessing the enteric nervous system (ENS). This study is focused on the ENS, vagus nerves and GI barrier in young MC v clean air controls. Electron microscopy of epithelial, endothelial and neural cells and immunoreactivity of stomach and vagus to phosphorylated ɑ-synuclein Ser129 and Hyperphosphorylated-Tau (Htau) were evaluated and CDNPs measured in ENS. CDNPs were abundant in erythrocytes, unmyelinated submucosal, perivascular and intramuscular nerve fibers, ganglionic neurons and vagus nerves and associated with organelle pathology. ɑSyn and Htau were present in 25/27 MC gastric,15/26 vagus and 18/27 gastric and 2/26 vagus samples respectively. We strongly suggest CDNPs are penetrating and damaging the GI barrier and reaching preganglionic parasympathetic fibers and the vagus nerve. This work highlights the potential role of CDNPs in the neuroenteric hyperphosphorylated ɑ-Syn and tau pathology as seen in Parkinson and Alzheimer's diseases. Highly oxidative, ubiquitous CDNPs constitute a biologically plausible path into Parkinson's and Alzheimer's pathogenesis.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- The University of Montana, Missoula, MT 59812, USA; Universidad del Valle de México, Mexico City 14370, Mexico.
| | | | | | | | | |
Collapse
|
21
|
Briceño MP, Nascimento LAC, Nogueira NP, Barenco PVC, Ferro EAV, Rezende-Oliveira K, Goulart LR, Alves PT, Barbosa BDF, Lima WR, Silva NM. Toxoplasma gondii Infection Promotes Epithelial Barrier Dysfunction of Caco-2 Cells. J Histochem Cytochem 2016; 64:459-69. [PMID: 27370796 DOI: 10.1369/0022155416656349] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/01/2016] [Indexed: 12/12/2022] Open
Abstract
After oral infection, Toxoplasma gondii invades intestinal cells, induces breakdown of intestinal physiology and barrier functions, and causes intestinal pathology in some animal species. Although parasites' invasion into host cells is a known phenomenon, the effects of T. gondii infection in the intestinal barrier are still not well established. To evaluate morphological and physiological modifications on the colorectal adenocarcinoma-derived Caco-2 cell line during T. gondii infection, microvilli, tight junction integrity, and transepithelial electrical resistance (TEER) were investigated under infection. It was observed that the dextran uptake (endocytosis) and distribution were smaller in infected than in noninfected Caco-2 cells. The infection leads to the partial loss of microvilli at the cell surface. Claudin-1, zonula occludens-1 (ZO-1), and occludin expressions were colocalized by immunofluorescence and presented discontinuous net patterns in infected cells. Immunoblotting analysis at 24 hr postinfection revealed decreasing expression of occludin and ZO-1 proteins, whereas claudin-1 presented similar expression level compared with noninfected cells. T. gondii decreased TEER in Caco-2 cells 24 hr after infection. Our results suggest that T. gondii infection may lead to the loss of integrity of intestinal mucosa, resulting in impaired barrier function.
Collapse
Affiliation(s)
- Marisol Pallete Briceño
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS)
| | | | - Nathalia Pires Nogueira
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS)
| | | | - Eloisa Amália Vieira Ferro
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS),Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil (BFB, EAVF)
| | - Karine Rezende-Oliveira
- Laboratory of Biomedical Sciences, Federal University of Uberlândia, Ituiutaba, Minas Gerais, Brazil (KR-O)
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil (LRG, PTA)
| | - Patrícia Terra Alves
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS),Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil (LRG, PTA)
| | - Bellisa de Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil (BFB, EAVF)
| | - Wânia Rezende Lima
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS),Institute of Exact and Natural Sciences, Federal University of Mato Grosso, Rondonópolis, Mato Grosso, Brazil (WRL)
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences (MPB, LACN, NPN, PVCB, WRL, NMS)
| |
Collapse
|
22
|
Hepatitis C virus depends on E-cadherin as an entry factor and regulates its expression in epithelial-to-mesenchymal transition. Proc Natl Acad Sci U S A 2016; 113:7620-5. [PMID: 27298373 DOI: 10.1073/pnas.1602701113] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatitis C virus (HCV) enters the host cell through interactions with a cascade of cellular factors. Although significant progress has been made in understanding HCV entry, the precise mechanisms by which HCV exploits the receptor complex and host machinery to enter the cell remain unclear. This intricate process of viral entry likely depends on additional yet-to-be-defined cellular molecules. Recently, by applying integrative functional genomics approaches, we identified and interrogated distinct sets of host dependencies in the complete HCV life cycle. Viral entry assays using HCV pseudoparticles (HCVpps) of various genotypes uncovered multiple previously unappreciated host factors, including E-cadherin, that mediate HCV entry. E-cadherin silencing significantly inhibited HCV infection in Huh7.5.1 cells, HepG2/miR122/CD81 cells, and primary human hepatocytes at a postbinding entry step. Knockdown of E-cadherin, however, had no effect on HCV RNA replication or internal ribosomal entry site (IRES)-mediated translation. In addition, an E-cadherin monoclonal antibody effectively blocked HCV entry and infection in hepatocytes. Mechanistic studies demonstrated that E-cadherin is closely associated with claudin-1 (CLDN1) and occludin (OCLN) on the cell membrane. Depletion of E-cadherin drastically diminished the cell-surface distribution of these two tight junction proteins in various hepatic cell lines, indicating that E-cadherin plays an important regulatory role in CLDN1/OCLN localization on the cell surface. Furthermore, loss of E-cadherin expression in hepatocytes is associated with HCV-induced epithelial-to-mesenchymal transition (EMT), providing an important link between HCV infection and liver cancer. Our data indicate that a dynamic interplay among E-cadherin, tight junctions, and EMT exists and mediates an important function in HCV entry.
Collapse
|
23
|
Mateo M, Generous A, Sinn PL, Cattaneo R. Connections matter--how viruses use cell–cell adhesion components. J Cell Sci 2016; 128:431-9. [PMID: 26046138 DOI: 10.1242/jcs.159400] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The epithelium is a highly organized type of animal tissue. Except for blood and lymph vessels, epithelial cells cover the body, line its cavities in single or stratified layers and support exchange between compartments. In addition, epithelia offer to the body a barrier to pathogen invasion. To transit through or to replicate in epithelia, viruses have to face several obstacles, starting from cilia and glycocalyx where they can be neutralized by secreted immunoglobulins. Tight junctions and adherens junctions also prevent viruses to cross the epithelial barrier. However, viruses have developed multiple strategies to blaze their path through the epithelium by utilizing components of cell–cell adhesion structures as receptors. In this Commentary, we discuss how viruses take advantage of the apical junction complex to spread. Whereas some viruses quickly disrupt epithelium integrity, others carefully preserve it and use cell adhesion proteins and their cytoskeletal connections to rapidly spread laterally. This is exemplified by the hidden transmission of enveloped viruses that use nectins as receptors. Finally, several viruses that replicate preferentially in cancer cells are currently used as experimental cancer therapeutics. Remarkably, these viruses use cell adhesion molecules as receptors, probably because--to reach tumors and metastases--ncolytic viruses must efficiently traverse or break epithelia.
Collapse
|
24
|
Askarian F, Ajayi C, Hanssen AM, van Sorge NM, Pettersen I, Diep DB, Sollid JUE, Johannessen M. The interaction between Staphylococcus aureus SdrD and desmoglein 1 is important for adhesion to host cells. Sci Rep 2016; 6:22134. [PMID: 26924733 PMCID: PMC4770587 DOI: 10.1038/srep22134] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/03/2016] [Indexed: 12/27/2022] Open
Abstract
Staphylococcus aureus is known as a frequent colonizer of the skin and mucosa. Among bacterial factors involved in colonization are adhesins such as the microbial surface components recognizing adhesive matrix molecules (MSCRAMMs). Serine aspartate repeat containing protein D (SdrD) is involved in adhesion to human squamous cells isolated from the nose. Here, we identify Desmoglein 1 (Dsg1) as a novel interaction partner for SdrD. Genetic deletion of sdrD in S. aureus NCTC8325-4 through allelic replacement resulted in decreased bacterial adherence to Dsg1- expressing HaCaT cells in vitro. Complementary gain-of-function was demonstrated by heterologous expression of SdrD in Lactococcus lactis, which increased adherence to HaCaT cells. Also ectopic expression of Dsg1 in HEK293 cells resulted in increased adherence of S. aureus NCTC8325-4 in vitro. Increased adherence of NCTC8325-4, compared to NCTC8325-4ΔsdrD, to the recombinant immobilized Dsg1 demonstrated direct interaction between SdrD and Dsg1. Specificity of SdrD interaction with Dsg1 was further verified using flow cytometry and confirmed binding of recombinant SdrD to HaCaT cells expressing Dsg1 on their surface. These data demonstrate that Dsg1 is a host ligand for SdrD.
Collapse
Affiliation(s)
- Fatemeh Askarian
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Clement Ajayi
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Anne-Merethe Hanssen
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Nina M van Sorge
- Medical Microbiology, University Medical Center Utrecht, Utrecht 3584CX, The Netherlands
| | - Ingvild Pettersen
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Dzung B Diep
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Science, Ås, Norway
| | - Johanna U E Sollid
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Mona Johannessen
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| |
Collapse
|
25
|
Porlan E, Martí-Prado B, Consiglio A, Fariñas I. Stable and Efficient Genetic Modification of Cells in the Adult Mouse V-SVZ for the Analysis of Neural Stem Cell Autonomous and Non-autonomous Effects. J Vis Exp 2016:53282. [PMID: 26967974 DOI: 10.3791/53282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Relatively quiescent somatic stem cells support life-long cell renewal in most adult tissues. Neural stem cells in the adult mammalian brain are restricted to two specific neurogenic niches: the subgranular zone of the dentate gyrus in the hippocampus and the ventricular-subventricular zone (V-SVZ; also called subependymal zone or SEZ) in the walls of the lateral ventricles. The development of in vivo gene transfer strategies for adult stem cell populations (i.e. those of the mammalian brain) resulting in long-term expression of desired transgenes in the stem cells and their derived progeny is a crucial tool in current biomedical and biotechnological research. Here, a direct in vivo method is presented for the stable genetic modification of adult mouse V-SVZ cells that takes advantage of the cell cycle-independent infection by LVs and the highly specialized cytoarchitecture of the V-SVZ niche. Specifically, the current protocol involves the injection of empty LVs (control) or LVs encoding specific transgene expression cassettes into either the V-SVZ itself, for the in vivo targeting of all types of cells in the niche, or into the lateral ventricle lumen, for the targeting of ependymal cells only. Expression cassettes are then integrated into the genome of the transduced cells and fluorescent proteins, also encoded by the LVs, allow the detection of the transduced cells for the analysis of cell autonomous and non-autonomous, niche-dependent effects in the labeled cells and their progeny.
Collapse
Affiliation(s)
- Eva Porlan
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO)
| | - Beatriz Martí-Prado
- Centro de Investigaciones Biomédicas en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Departmento de Biologìa Celular, Universidad de Valencia
| | - Antonella Consiglio
- Institut de Biomedicina de la Universitat de Barcelona (IBUB); Department of Molecular and Translational Medicine, Fibroblast Reprogramming Unit, University of Brescia;
| | - Isabel Fariñas
- Centro de Investigaciones Biomédicas en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Departmento de Biologìa Celular, Universidad de Valencia;
| |
Collapse
|
26
|
Abstract
The currently available somatostatin receptor ligands (SRLs) and growth hormone (GH) antagonists are used to control levels of GH and insulin-like growth factor 1 (IGF-1) in patients with acromegaly. However, these therapies are limited by wide variations in efficacy, associated adverse effects and the need for frequent injections. A phase III trial of oral octreotide capsules demonstrated that this treatment can safely sustain suppressed levels of GH and IGF-1 and reduce the severity of symptoms in patients with acromegaly previously controlled by injectable SRL therapy, with the added benefit of no injection-site reactions. Phase I and phase II trials of the pan-selective SRL DG3173, the liquid crystal octreotide depot CAM2029 and an antisense oligonucleotide directed against the GH receptor have shown that these agents can be used to achieve biochemical suppression in acromegaly and have favourable safety profiles. This Review outlines the need for new therapeutic agents for patients with acromegaly, reviews clinical trial data of investigational agents and considers how these therapies might best be integrated into clinical practice.
Collapse
Affiliation(s)
- Shlomo Melmed
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Room 2015, Los Angeles, California 90048, USA
| |
Collapse
|
27
|
Fernandes LG, Siqueira GH, Teixeira ARF, Silva LP, Figueredo JM, Cosate MR, Vieira ML, Nascimento ALTO. Leptospira spp.: Novel insights into host-pathogen interactions. Vet Immunol Immunopathol 2015; 176:50-7. [PMID: 26727033 DOI: 10.1016/j.vetimm.2015.12.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 01/30/2023]
Abstract
Leptospirosis is a widespread zoonosis caused by pathogenic Leptospira spp. It is an important infectious disease that affects humans and animals. The disease causes economic losses as it affects livestock, with decreased milk production and death. Our group is investigating the genome sequences of L. interrogans targeting surface-exposed proteins because, due to their location, these proteins are capable to interact with several host components that could allow establishment of the infection. These interactions may involve adhesion of the bacteria to extracellular matrix (ECM) components and, hence, help bacterial colonization. The bacteria could also react with the host fibrinolytic system and/or with the coagulation cascade components, such as, plasminogen (PLG) and fibrinogen (Fg), respectively. The binding with the first system generates plasmin (PLA), increasing the proteolytic power of the bacteria, while the second interferes with clotting in a thrombin-catalyzed reaction, which may promote hemorrhage foci and increase bacterial dissemination. Interaction with the complement system negative regulators may help bacteria to evade the host immune system, facilitating the invasion. This work compiles the main described leptospiral proteins that could act as adhesins, as PLG and fibrinogen receptors and as complement regulator binding proteins. We present models in which we suggest possible mechanisms of how leptospires might colonize and invade host tissues, causing the disease. Understanding leptospiral pathogenesis will help to identify antigen candidates that would contribute to the development of more effective vaccines and diagnostic tests.
Collapse
Affiliation(s)
- Luis G Fernandes
- Centro de Biotecnologia, Instituto Butantan, Avenida Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Programa de Pós Graduação Interunidades em Biotecnologia, Instituto de Ciências Biomédicas, USP, Avenida Prof. Lineu Prestes, 1730, 05508-900, São Paulo, SP, Brazil
| | - Gabriela H Siqueira
- Centro de Biotecnologia, Instituto Butantan, Avenida Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Programa de Pós Graduação Interunidades em Biotecnologia, Instituto de Ciências Biomédicas, USP, Avenida Prof. Lineu Prestes, 1730, 05508-900, São Paulo, SP, Brazil
| | - Aline R F Teixeira
- Centro de Biotecnologia, Instituto Butantan, Avenida Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Programa de Pós Graduação Interunidades em Biotecnologia, Instituto de Ciências Biomédicas, USP, Avenida Prof. Lineu Prestes, 1730, 05508-900, São Paulo, SP, Brazil
| | - Lucas P Silva
- Centro de Biotecnologia, Instituto Butantan, Avenida Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Programa de Pós Graduação Interunidades em Biotecnologia, Instituto de Ciências Biomédicas, USP, Avenida Prof. Lineu Prestes, 1730, 05508-900, São Paulo, SP, Brazil
| | - Jupciana M Figueredo
- Centro de Biotecnologia, Instituto Butantan, Avenida Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Programa de Pós Graduação Interunidades em Biotecnologia, Instituto de Ciências Biomédicas, USP, Avenida Prof. Lineu Prestes, 1730, 05508-900, São Paulo, SP, Brazil
| | - Maria R Cosate
- Centro de Biotecnologia, Instituto Butantan, Avenida Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Monica L Vieira
- Centro de Biotecnologia, Instituto Butantan, Avenida Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | - Ana L T O Nascimento
- Centro de Biotecnologia, Instituto Butantan, Avenida Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil; Programa de Pós Graduação Interunidades em Biotecnologia, Instituto de Ciências Biomédicas, USP, Avenida Prof. Lineu Prestes, 1730, 05508-900, São Paulo, SP, Brazil.
| |
Collapse
|
28
|
Weight CM, Jones EJ, Horn N, Wellner N, Carding SR. Elucidating pathways of Toxoplasma gondii invasion in the gastrointestinal tract: involvement of the tight junction protein occludin. Microbes Infect 2015; 17:698-709. [PMID: 26183539 DOI: 10.1016/j.micinf.2015.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/17/2015] [Accepted: 07/02/2015] [Indexed: 01/28/2023]
Abstract
Toxoplasma gondii is an obligate intracellular parasite infecting one third of the world's population. The small intestine is the parasite's primary route of infection, although the pathway of epithelium transmigration remains unclear. Using an in vitro invasion assay and live imaging we showed that T. gondii (RH) tachyzoites infect and transmigrate between adjacent intestinal epithelial cells in polarized monolayers without altering barrier integrity, despite eliciting the production of specific inflammatory mediators and chemokines. During invasion, T. gondii co-localized with occludin. Reducing the levels of endogenous cellular occludin with specific small interfering RNAs significantly reduced the ability of T. gondii to penetrate between and infect epithelial cells. Furthermore, an in vitro invasion and binding assays using recombinant occludin fragments established the capacity of the parasite to bind occludin and in particular to the extracellular loops of the protein. These findings provide evidence for occludin playing a role in the invasion of T. gondii in small intestinal epithelial cells.
Collapse
Affiliation(s)
- Caroline M Weight
- Gut Health and Food Safety Institute Strategic Programme, Norwich Research Park, Norwich, NR4 7UA, UK; Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7UA, UK
| | - Emily J Jones
- Gut Health and Food Safety Institute Strategic Programme, Norwich Research Park, Norwich, NR4 7UA, UK; Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7UA, UK
| | - Nikki Horn
- Gut Health and Food Safety Institute Strategic Programme, Norwich Research Park, Norwich, NR4 7UA, UK
| | - Nikolaus Wellner
- Analytical Sciences Unit, Institute of Food Research, Norwich Research Park, Norwich, NR4 7UA, UK
| | - Simon R Carding
- Gut Health and Food Safety Institute Strategic Programme, Norwich Research Park, Norwich, NR4 7UA, UK; Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7UA, UK.
| |
Collapse
|
29
|
Short-term effects of Poly(I:C) on gut permeability. Pharmacol Res 2015; 101:130-6. [PMID: 26145280 DOI: 10.1016/j.phrs.2015.06.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 06/25/2015] [Accepted: 06/25/2015] [Indexed: 02/07/2023]
Abstract
The intestinal barrier function depends on an adequate response to pathogens by the epithelium. Toll-like receptor 3 (TLR-3) recognizes double-stranded RNA, a virus-associated molecular pattern. Activation of TLR-3 with Poly(I:C), a synthetic agonist, modulates tissue repair and permeability in other epithelia; however, the effects of local luminal TLR-3 agonists on gut barrier function are unknown. The aim of this investigation was to evaluate short-term effects of Poly(I:C) on rat ileal and colonic permeability ex vivo. We also studied the acute effects of intrarectal administration of Poly(I:C) on colonic barrier function. Ileum tissues displayed decreased transepithelial electrical resistance (TEER) 1h after incubation with 200μg/mL Poly(I:C); however, the mucosa-to-serosa transit of macromolecules (4.4 and 40kDa dextrans - TD4.4 and FD40, respectively) remained unchanged. Conversely, colon tissue preparations stimulated with 200μg/mL Poly(I:C) showed a decreased thinning of the mucosal layer after 2h and a decreased transit of FD40 after 3h, in comparison to controls. There was no change in colonic TEER after 3h of treatment. In addition, colon tissue taken from rats 6h after an intrarectal administration of 100μg Poly(I:C) also showed decreased permeability to FD40 in the everted gut sac assay at 3h post-extraction. Tissue morphology remained unchanged. Our results suggest that an acute exposure to Poly(I:C) reduces colon permeability to macromolecules but increases ileum permeability to electrolytes/small molecules ex vivo. Although the mechanism associated to these effects needs further investigation, to our knowledge this is the first report of a direct effect of a TLR-3 ligand in intestinal barrier function and may be of significance to understand region-specific interactions between gut mucosa and microbiota.
Collapse
|
30
|
Barrile R, Kasendra M, Rossi-Paccani S, Merola M, Pizza M, Baldari C, Soriani M, Aricò B. Neisseria meningitidis subverts the polarized organization and intracellular trafficking of host cells to cross the epithelial barrier. Cell Microbiol 2015; 17:1365-75. [PMID: 25801707 DOI: 10.1111/cmi.12439] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 03/13/2015] [Accepted: 03/14/2015] [Indexed: 12/19/2022]
Abstract
Translocation of the nasopharyngeal barrier by Neisseria meningitidis occurs via an intracellular microtubule-dependent pathway and represents a crucial step in its pathogenesis. Despite this fact, the interaction of invasive meningococci with host subcellular compartments and the resulting impact on their organization and function have not been investigated. The influence of serogroup B strain MC58 on host cell polarity and intracellular trafficking system was assessed by confocal microscopy visualization of different plasma membrane-associated components (such as E-cadherin, ZO-1 and transferrin receptor) and evaluation of the transferrin uptake and recycling in infected Calu-3 monolayers. Additionally, the association of N. meningitidis with different endosomal compartments was evaluated through the concomitant staining of bacteria and markers specific for Rab11, Rab22a, Rab25 and Rab3 followed by confocal microscopy imaging. Subversion of the host cell architecture and intracellular trafficking system, denoted by mis-targeting of cell plasma membrane components and perturbations of transferrin transport, was shown to occur in response to N. meningitidis infection. Notably, the appearance of all of these events seems to positively correlate with the efficiency of N. meningitidis to cross the epithelial barrier. Our data reveal for the first time that N. meningitidis is able to modulate the host cell architecture and function, which might serve as a strategy of this pathogen for overcoming the nasopharyngeal barrier without affecting the monolayer integrity.
Collapse
Affiliation(s)
- Riccardo Barrile
- Department of Microbial Molecular Biology, Novartis Vaccines and Diagnostics (a GSK company), Siena, Italy.,Biomimetic Microsystems platform, Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Magdalena Kasendra
- Department of Microbial Molecular Biology, Novartis Vaccines and Diagnostics (a GSK company), Siena, Italy
| | - Silvia Rossi-Paccani
- Department of Microbial Molecular Biology, Novartis Vaccines and Diagnostics (a GSK company), Siena, Italy
| | - Marcello Merola
- Department of Microbial Molecular Biology, Novartis Vaccines and Diagnostics (a GSK company), Siena, Italy.,Department of Biology, University of Naples 'Federico II', Napoli, Italy
| | - Mariagrazia Pizza
- Department of Microbial Molecular Biology, Novartis Vaccines and Diagnostics (a GSK company), Siena, Italy
| | - Cosima Baldari
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Marco Soriani
- Department of Microbial Molecular Biology, Novartis Vaccines and Diagnostics (a GSK company), Siena, Italy
| | - Beatrice Aricò
- Department of Microbial Molecular Biology, Novartis Vaccines and Diagnostics (a GSK company), Siena, Italy
| |
Collapse
|
31
|
Induction of size-dependent breakdown of blood-milk barrier in lactating mice by TiO2 nanoparticles. PLoS One 2015; 10:e0122591. [PMID: 25849145 PMCID: PMC4388820 DOI: 10.1371/journal.pone.0122591] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 02/11/2015] [Indexed: 11/19/2022] Open
Abstract
This study aims to investigate the potential nanotoxic effects of TiO2 nanoparticles (TNPs) to dams and pups during lactation period. TiO2 nanoparticles are accumulated in mammary glands of lactating mice after i.v. administration. This accumulation of TiO2 NP likely causes a ROS-induced disruption of tight junction of the blood-milk barrier as indicated by the loss of tight junction proteins and the shedding of alveolar epithelial cells. Compared to larger TNPs (50 nm), smaller ones (8 nm) exhibit a higher accumulation in mammary glands and are more potent in causing perturbations to blood-milk barrier. An alarming finding is that the smaller TNPs (8 nm) are transferred from dams to pups through breastfeeding, likely through the disrupted blood-milk barrier. However, during the lactation period, the nutrient quality of milk from dams and the early developmental landmarks of the pups are not affected by above perturbations.
Collapse
|
32
|
Dibao-Dina A, Follet J, Ibrahim M, Vlandas A, Senez V. Electrical impedance sensor for quantitative monitoring of infection processes on HCT-8 cells by the waterborne parasite Cryptosporidium. Biosens Bioelectron 2015; 66:69-76. [DOI: 10.1016/j.bios.2014.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/28/2014] [Accepted: 11/06/2014] [Indexed: 01/07/2023]
|
33
|
Ribet D, Cossart P. How bacterial pathogens colonize their hosts and invade deeper tissues. Microbes Infect 2015; 17:173-83. [PMID: 25637951 DOI: 10.1016/j.micinf.2015.01.004] [Citation(s) in RCA: 470] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 01/18/2015] [Accepted: 01/19/2015] [Indexed: 02/06/2023]
Abstract
Bacterial pathogens have evolved a wide range of strategies to colonize and invade human organs, despite the presence of multiple host defense mechanisms. In this review, we will describe how pathogenic bacteria can adhere and multiply at the surface of host cells, how some bacteria can enter and proliferate inside these cells, and finally how pathogens may cross epithelial or endothelial host barriers and get access to internal tissues, leading to severe diseases in humans.
Collapse
Affiliation(s)
- David Ribet
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Département de Biologie Cellulaire et Infection, F-75015 Paris, France; INSERM, U604, F-75015 Paris, France; INRA, USC2020, F-75015 Paris, France.
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Département de Biologie Cellulaire et Infection, F-75015 Paris, France; INSERM, U604, F-75015 Paris, France; INRA, USC2020, F-75015 Paris, France.
| |
Collapse
|
34
|
Abstract
It is now emerging that a number of cellular targets of pathogens are involved in the establishment and/or maintenance of epithelial cell polarity. Increasing evidence also suggests that cancer-causing pathogens such as Helicobacter pylori (H. pylori) and human papilloma virus (HPV) may induce oncogenesis by disrupting cell polarity. This is mainly achieved through their ability to deregulate the function of cell polarity components and/or regulators. Hence cell polarity represents the first line of defence against infection. Interestingly, EGFR/RAS oncogenic signals also induce cancer cell invasion by inducing epithelial to mesenchymal transition (EMT). Since the loss of cell polarity is a prerequisition of EMT, cell polarity also represents the last line of defence against cancer cell invasion. As such we argue that cell polarity may be a key defence mechanism against infection and cancer cell invasion. The potential role of cell polarity as a gatekeeper against cancer through its ability to regulate asymmetric cell division and tumour suppression has been discussed in a number of recent reviews. In this review we will focus on the role of cell polarity as a potential target of infection and cancer cell invasion.
Collapse
Affiliation(s)
- Klaus Ebnet
- grid.5949.10000000121729288Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University Münster, Münster, Nordrhein-Westfalen Germany
| | | |
Collapse
|
35
|
Abstract
Technological advances in the large scale analysis of human genetics have generated profound insights into possible genetic contributions to chronic diseases including the inflammatory bowel diseases (IBDs), Crohn's disease and ulcerative colitis. To date, 163 distinct genetic risk loci have been associated with either Crohn's disease or ulcerative colitis, with a substantial degree of genetic overlap between these 2 conditions. Although many risk variants show a reproducible correlation with disease, individual gene associations only affect a subset of patients, and the functional contribution(s) of these risk variants to the onset of IBD is largely undetermined. Although studies in twins have demonstrated that the development of IBD is not mediated solely by genetic risk, it is nevertheless important to elucidate the functional consequences of risk variants for gene function in relevant cell types known to regulate key physiological processes that are compromised in IBD. This article will discuss IBD candidate genes that are known to be, or are suspected of being, involved in regulating the intestinal epithelial barrier and several of the physiological processes presided over by this dynamic and versatile layer of cells. This will include assembly and regulation of tight junctions, cell adhesion and polarity, mucus and glycoprotein regulation, bacterial sensing, membrane transport, epithelial differentiation, and restitution.
Collapse
|
36
|
Wu Z, Mirza H, Tan KSW. Intra-subtype variation in enteroadhesion accounts for differences in epithelial barrier disruption and is associated with metronidazole resistance in Blastocystis subtype-7. PLoS Negl Trop Dis 2014; 8:e2885. [PMID: 24851944 PMCID: PMC4031124 DOI: 10.1371/journal.pntd.0002885] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 04/08/2014] [Indexed: 01/13/2023] Open
Abstract
Blastocystis is an extracellular, enteric pathogen that induces intestinal disorders in a range of hosts including humans. Recent studies have identified potential parasite virulence factors in and host responses to this parasite; however, little is known about Blastocystis-host attachment, which is crucial for colonization and virulence of luminal stages. By utilizing 7 different strains of the parasite belonging to two clinically relevant subtypes ST-4 and ST-7, we investigated Blastocystis-enterocyte adhesion and its association with parasite-induced epithelial barrier disruption. We also suggest that drug resistance in ST-7 strains might result in fitness cost that manifested as impairment of parasite adhesion and, consequently, virulence. ST-7 parasites were generally highly adhesive to Caco-2 cells and preferred binding to intercellular junctions. These strains also induced disruption of ZO-1 and occludin tight junction proteins as well as increased dextran-FITC flux across epithelial monolayers. Interestingly, their adhesion was correlated with metronidazole (Mz) susceptibility. Mz resistant (Mzr) strains were found to be less pathogenic, owing to compromised adhesion. Moreover, tolerance of nitrosative stress was also reduced in the Mzr strains. In conclusion, the findings indicate that Blastocystis attaches to intestinal epithelium and leads to epithelial barrier dysfunction and that drug resistance might entail a fitness cost in parasite virulence by limiting entero-adhesiveness. This is the first study of the cellular basis for strain-to-strain variation in parasite pathogenicity. Intra- and inter-subtype variability in cytopathogenicity provides a possible explanation for the diverse clinical outcomes of Blastocystis infections.
Collapse
Affiliation(s)
- Zhaona Wu
- Laboratory of Molecular and Cellular Parasitology, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haris Mirza
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Kevin Shyong Wei Tan
- Laboratory of Molecular and Cellular Parasitology, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
37
|
Losa D, Köhler T, Bellec J, Dudez T, Crespin S, Bacchetta M, Boulanger P, Hong SS, Morel S, Nguyen TH, van Delden C, Chanson M. Pseudomonas aeruginosa–Induced Apoptosis in Airway Epithelial Cells Is Mediated by Gap Junctional Communication in a JNK-Dependent Manner. THE JOURNAL OF IMMUNOLOGY 2014; 192:4804-12. [DOI: 10.4049/jimmunol.1301294] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
38
|
McCormack J, Welsh NJ, Braga VMM. Cycling around cell-cell adhesion with Rho GTPase regulators. J Cell Sci 2014; 126:379-91. [PMID: 23547086 DOI: 10.1242/jcs.097923] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The formation and stability of epithelial adhesive systems, such as adherens junctions, desmosomes and tight junctions, rely on a number of cellular processes that ensure a dynamic interaction with the cortical cytoskeleton, and appropriate delivery and turnover of receptors at the surface. Unique signalling pathways must be coordinated to allow the coexistence of distinct adhesive systems at discrete sub-domains along junctions and the specific properties they confer to epithelial cells. Rho, Rac and Cdc42 are members of the Rho small GTPase family, and are well-known regulators of cell-cell adhesion. The spatio-temporal control of small GTPase activation drives specific intracellular processes to enable the hierarchical assembly, morphology and maturation of cell-cell contacts. Here, we discuss the small GTPase regulators that control the precise amplitude and duration of the levels of active Rho at cell-cell contacts, and the mechanisms that tailor the output of Rho signalling to a particular cellular event. Interestingly, the functional interaction is reciprocal; Rho regulators drive the maturation of cell-cell contacts, whereas junctions can also modulate the localisation and activity of Rho regulators to operate in diverse processes in the epithelial differentiation programme.
Collapse
Affiliation(s)
- Jessica McCormack
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London. Sir Alexander Fleming Building, London SW7 2AZ, UK
| | | | | |
Collapse
|
39
|
Zeldovich VB, Clausen CH, Bradford E, Fletcher DA, Maltepe E, Robbins JR, Bakardjiev AI. Placental syncytium forms a biophysical barrier against pathogen invasion. PLoS Pathog 2013; 9:e1003821. [PMID: 24348256 PMCID: PMC3861541 DOI: 10.1371/journal.ppat.1003821] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 10/11/2013] [Indexed: 02/02/2023] Open
Abstract
Fetal syncytiotrophoblasts form a unique fused multinuclear surface that is bathed in maternal blood, and constitutes the main interface between fetus and mother. Syncytiotrophoblasts are exposed to pathogens circulating in maternal blood, and appear to have unique resistance mechanisms against microbial invasion. These are due in part to the lack of intercellular junctions and their receptors, the Achilles heel of polarized mononuclear epithelia. However, the syncytium is immune to receptor-independent invasion as well, suggesting additional general defense mechanisms against infection. The difficulty of maintaining and manipulating primary human syncytiotrophoblasts in culture makes it challenging to investigate the cellular and molecular basis of host defenses in this unique tissue. Here we present a novel system to study placental pathogenesis using murine trophoblast stem cells (mTSC) that can be differentiated into syncytiotrophoblasts and recapitulate human placental syncytium. Consistent with previous results in primary human organ cultures, murine syncytiotrophoblasts were found to be resistant to infection with Listeria monocytogenes via direct invasion and cell-to-cell spread. Atomic force microscopy of murine syncytiotrophoblasts demonstrated that these cells have a greater elastic modulus than mononuclear trophoblasts. Disruption of the unusually dense actin structure--a diffuse meshwork of microfilaments--with Cytochalasin D led to a decrease in its elastic modulus by 25%. This correlated with a small but significant increase in invasion of L. monocytogenes into murine and human syncytium. These results suggest that the syncytial actin cytoskeleton may form a general barrier against pathogen entry in humans and mice. Moreover, murine TSCs are a genetically tractable model system for the investigation of specific pathways in syncytial host defenses.
Collapse
Affiliation(s)
- Varvara B. Zeldovich
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
- Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, California, United States of America
| | - Casper H. Clausen
- Department of Bioengineering and Program in Biophysics, University of California, Berkeley, Berkeley, California, United States of America
| | - Emily Bradford
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
- Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, California, United States of America
- Biomedical Sciences Program, University of California, San Francisco, San Francisco, California, United States of America
| | - Daniel A. Fletcher
- Department of Bioengineering and Program in Biophysics, University of California, Berkeley, Berkeley, California, United States of America
| | - Emin Maltepe
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
- Biomedical Sciences Program, University of California, San Francisco, San Francisco, California, United States of America
| | - Jennifer R. Robbins
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
- Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, California, United States of America
- Department of Biology, Xavier University, Cincinnati, Ohio, United States of America
| | - Anna I. Bakardjiev
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, United States of America
- Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, California, United States of America
- Biomedical Sciences Program, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Kasendra M, Barrile R, Leuzzi R, Soriani M. Clostridium difficile toxins facilitate bacterial colonization by modulating the fence and gate function of colonic epithelium. J Infect Dis 2013; 209:1095-104. [PMID: 24273043 DOI: 10.1093/infdis/jit617] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The contribution of Clostridium difficile toxin A and B (TcdA and TcdB) to cellular intoxication has been studied extensively, but their impact on bacterial colonization remains unclear. By setting up 2- and 3-dimensional in vitro models of polarized gut epithelium, we investigated how C. difficile infection is affected by host cell polarity and whether TcdA and TcdB contribute to such events. Indeed, we observed that C. difficile adhesion and penetration of the mucosal barrier are substantially enhanced in poorly polarized or ethylene glycol tetraacetic acid-treated cells, indicating that bacteria bind preferentially to the basolateral (BL) cell surface. In this context, we demonstrated that sub-lethal concentrations of C. difficile TcdA are able to alter cell polarity by causing redistribution of plasma membrane components between distinct surface domains. Taken together, the data suggest that toxin-mediated modulation of host cell organization may account for the capacity of this opportunistic pathogen to gain access to BL receptors, leading to a successful colonization of the colonic mucosa.
Collapse
|
41
|
Padmashali R, You H, Karnik N, Lei P, Andreadis ST. Adherens junction formation inhibits lentivirus entry and gene transfer. PLoS One 2013; 8:e79265. [PMID: 24236116 PMCID: PMC3827380 DOI: 10.1371/journal.pone.0079265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/27/2013] [Indexed: 01/04/2023] Open
Abstract
Although cellular signaling pathways that affect lentivirus infection have been investigated, the role of cell-cell interactions in lentiviral gene delivery remains elusive. In the course of our studies we observed that lentiviral gene transfer was a strong function of the position of epithelial cells within colonies. While peripheral cells were transduced efficiently, cells in the center of colonies were resistant to gene transfer. In addition, gene delivery was enhanced significantly under culture conditions that disrupted adherens junctions (AJ) but decreased upon AJ formation. In agreement, gene knockdown and gain-of-function approaches showed that α-catenin, a key component of the AJ complex prevented lentivirus gene transfer. Using a doxycycline regulatable system we showed that expression of dominant negative E-cadherin enhanced gene transfer in a dose-dependent manner. In addition, dissolution of AJ by doxycycline increased entry of lentiviral particles into the cell cytoplasm in a dose-dependent manner. Taken together our results demonstrate that AJ formation renders cells non-permissive to lentiviral gene transfer and may facilitate development of simple means to enhance gene delivery or combat virus infection.
Collapse
Affiliation(s)
- Roshan Padmashali
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, New York, United States of America
| | - Hui You
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, New York, United States of America
| | - Nikhila Karnik
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, New York, United States of America
| | - Pedro Lei
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, New York, United States of America
| | - Stelios T. Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, New York, United States of America
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Amherst, New York, United States of America
- Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
42
|
Humphries AC, Way M. The non-canonical roles of clathrin and actin in pathogen internalization, egress and spread. Nat Rev Microbiol 2013; 11:551-60. [PMID: 24020073 DOI: 10.1038/nrmicro3072] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The role of clathrin in pathogen entry has received much attention and has highlighted the adaptability of clathrin during internalization. Recent studies have now uncovered additional roles for clathrin and have put the spotlight on its role in pathogen spread. Here, we discuss the manipulation of clathrin by pathogens, with specific attention to the processes that occur at the plasma membrane. In the majority of cases, both clathrin and the actin cytoskeleton are hijacked, so we also examine the interplay between these two systems and their role during pathogen internalization, egress and spread.
Collapse
Affiliation(s)
- Ashley C Humphries
- Cancer Research UK London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | | |
Collapse
|
43
|
Escherichia coli heat-stable toxin b impairs intestinal epithelial barrier function by altering tight junction proteins. Infect Immun 2013; 81:2819-27. [PMID: 23716609 DOI: 10.1128/iai.00455-13] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Escherichia coli heat-stable toxin b (STb) causes diarrhea in animals. STb binds to sulfatide, its receptor, and is then internalized. In the cytoplasm, through a cascade of events, STb triggers the opening of ion channels, allowing ion secretion and water loss and leading to diarrhea. Tight junctions (TJs) are well known for controlling paracellular traffic of ions and water by forming a physical intercellular barrier in epithelial cells, and some bacterial toxins are known to affect adversely TJs. The present study aimed at determining the effect of STb on TJs. T84 cells were treated for 24 h with purified STb and a nontoxic STb mutant (D30V). Transepithelial resistance (TER), paracellular flux marker, and confocal microscopy were used to analyze the effect of STb on TJs. Purified STb caused a significant reduction of TER parallel to an increase in paracellular permeability compared to the results seen in untreated cells or mutant D30V. The increased paracellular permeability was associated with a marked alteration of F-actin stress fibers. F-actin filament dissolution and condensation were accompanied by redistribution and/or fragmentation of ZO-1, claudin-1, and occludin. These changes were also observed following treatment of T84 cells with an 8-amino-acid peptide found in the STb sequence corresponding to a consensus sequence of Vibrio cholerae Zot toxin. These effects were not observed with a scrambled peptide or mutant D30V. Our findings indicate that STb induces epithelial barrier dysfunction through changes in TJ proteins that could contribute to diarrhea.
Collapse
|
44
|
Kobayashi K, Oyama S, Numata A, Rahman MM, Kumura H. Lipopolysaccharide disrupts the milk-blood barrier by modulating claudins in mammary alveolar tight junctions. PLoS One 2013; 8:e62187. [PMID: 23626786 PMCID: PMC3633878 DOI: 10.1371/journal.pone.0062187] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 03/19/2013] [Indexed: 12/13/2022] Open
Abstract
Mastitis, inflammation of the mammary gland, is the most costly common disease in the dairy industry, and is caused by mammary pathogenic bacteria, including Escherichia coli. The bacteria invade the mammary alveolar lumen and disrupt the blood-milk barrier. In normal mammary gland, alveolar epithelial tight junctions (TJs) contribute the blood-milk barrier of alveolar epithelium by blocking the leakage of milk components from the luminal side into the blood serum. In this study, we focused on claudin subtypes that participate in the alveolar epithelial TJs, because the composition of claudins is an important factor that affects TJ permeability. In normal mouse lactating mammary glands, alveolar TJs consist of claudin-3 without claudin-1, -4, and -7. In lipopolysaccharide (LPS)-induced mastitis, alveolar TJs showed 2-staged compositional changes in claudins. First, a qualitative change in claudin-3, presumably caused by phosphorylation and participation of claudin-7 in alveolar TJs, was recognized in parallel with the leakage of fluorescein isothiocyanate-conjugated albumin (FITC-albumin) via the alveolar epithelium. Second, claudin-4 participated in alveolar TJs with claudin-3 and claudin-7 12 h after LPS injection. The partial localization of claudin-1 was also observed by immunostaining. Coinciding with the second change of alveolar TJs, the severe disruption of the blood-milk barrier was recognized by ectopic localization of β-casein and much leakage of FITC-albumin. Furthermore, the localization of toll-like receptor 4 (TLR4) on the luminal side and NFκB activation by LPS was observed in the alveolar epithelial cells. We suggest that the weakening and disruption of the blood-milk barrier are caused by compositional changes of claudins in alveolar epithelial TJs through LPS/TLR4 signaling.
Collapse
Affiliation(s)
- Ken Kobayashi
- Laboratory of Dairy Food Science, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan.
| | | | | | | | | |
Collapse
|
45
|
Conti A, Sueur C, Lupo J, Brazzolotto X, Burmeister WP, Manet E, Gruffat H, Morand P, Boyer V. Interaction of Ubinuclein-1, a nuclear and adhesion junction protein, with the 14-3-3 epsilon protein in epithelial cells: implication of the PKA pathway. Eur J Cell Biol 2013; 92:105-11. [PMID: 23395486 DOI: 10.1016/j.ejcb.2012.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 12/19/2012] [Accepted: 12/23/2012] [Indexed: 10/27/2022] Open
Abstract
Ubinuclein-1 is a NACos (Nuclear and Adhesion junction Complex components) protein which shuttles between the nucleus and tight junctions, but its function in the latter is not understood. Here, by co-immunoprecipitation and confocal analysis, we show that Ubinuclein-1 interacts with the 14-3-3ɛ protein both in HT29 colon cells, and AGS gastric cells. This interaction is mediated by an Ubinuclein-1 phosphoserine motif. We show that the arginine residues (R56, R60 and R132) which form the 14-3-3ɛ ligand binding site are responsible for the binding of 14-3-3ɛ to phosphorylated Ubinuclein-1. Furthermore, we demonstrate that in vitro Ubinuclein-1 can be directly phosphorylated by cAMP-dependent protein kinase A. This in vitro phosphorylation allows binding of wildtype 14-3-3ɛ. Moreover, treatment of the cells with inhibitors of the cAMP-dependent protein kinase, KT5720 or H89, modifies the subcellular localization of Ubinuclein-1. Indeed, KT5720 and H89 greatly increase the staining of Ubinuclein-1 at the tight junctions in AGS gastric cells. In the presence of the kinase inhibitor KT5720, the amount of Ubinuclein-1 in the NP40 insoluble fraction is increased, together with actin. Moreover, treatment of the cells with KT5720 or H89 induces the concentration of Ubinuclein-1 at tricellular intersections of MDCK cells. Taken together, our findings demonstrate novel cell signaling trafficking by Ubinuclein-1 via association with 14-3-3ɛ following Ubinuclein-1 phosphorylation by the cAMP-dependent protein kinase-A.
Collapse
Affiliation(s)
- Audrey Conti
- Unit of Virus Host Cell Interactions (UVHCI), UJF Grenoble1-EMBL-CNRS UMI 3265, 6 rue Jules Horowitz, BP 181, F-38042 Grenoble Cedex 9, France
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Burlet E, Jain SK. Manganese supplementation reduces high glucose-induced monocyte adhesion to endothelial cells and endothelial dysfunction in Zucker diabetic fatty rats. J Biol Chem 2013; 288:6409-16. [PMID: 23329836 DOI: 10.1074/jbc.m112.447805] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Endothelial dysfunction is a hallmark of increased vascular inflammation, dyslipidemia, and the development of atherosclerosis in diabetes. Previous studies have reported lower levels of Mn(2+) in the plasma and lymphocytes of diabetic patients and in the heart and aortic tissue of patients with atherosclerosis. This study examines the hypothesis that Mn(2+) supplementation can reduce the markers/risk factors of endothelial dysfunction in type 2 diabetes. Human umbilical vein endothelial cells (HUVECs) were cultured with or without Mn(2+) supplementation and then exposed to high glucose (HG, 25 mm) to mimic diabetic conditions. Mn(2+) supplementation caused a reduction in monocyte adhesion to HUVECs treated with HG or MCP-1. Mn(2+) also inhibited ROS levels, MCP-1 secretion, and ICAM-1 up-regulation in HUVECs treated with HG. Silencing studies using siRNA against MnSOD showed that similar results were observed in MnSOD knockdown HUVECs following Mn(2+) supplementation, suggesting that the effect of manganese on monocyte adhesion to endothelial cells is mediated by ROS and ICAM-1, but not MnSOD. To validate the relevance of our findings in vivo, Zucker diabetic fatty rats were gavaged daily with water (placebo) or MnCl2 (16 mg/kg of body weight) for 7 weeks. When compared with placebo, Mn(2+)-supplemented rats showed lower blood levels of ICAM-1 (17%, p < 0.04), cholesterol (25%, p < 0.05), and MCP-1 (28%, p = 0.25). These in vitro and in vivo studies demonstrate that Mn(2+) supplementation can down-regulate ICAM-1 expression and ROS independently of MnSOD, leading to a decrease in monocyte adhesion to endothelial cells, and therefore can lower the risk of endothelial dysfunction in diabetes.
Collapse
Affiliation(s)
- Elodie Burlet
- Department of Pediatrics and Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA
| | | |
Collapse
|
47
|
Kunisawa J, Kiyono H. Immune regulation and monitoring at the epithelial surface of the intestine. Drug Discov Today 2013; 18:87-92. [DOI: 10.1016/j.drudis.2012.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 07/06/2012] [Accepted: 08/02/2012] [Indexed: 02/08/2023]
|
48
|
Saito M, Tucker DK, Kohlhorst D, Niessen CM, Kowalczyk AP. Classical and desmosomal cadherins at a glance. J Cell Sci 2012; 125:2547-52. [PMID: 22833291 DOI: 10.1242/jcs.066654] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Masataka Saito
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
49
|
Bonazzi M, Kühbacher A, Toledo-Arana A, Mallet A, Vasudevan L, Pizarro-Cerdá J, Brodsky FM, Cossart P. A common clathrin-mediated machinery co-ordinates cell-cell adhesion and bacterial internalization. Traffic 2012; 13:1653-66. [PMID: 22984946 DOI: 10.1111/tra.12009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 09/12/2012] [Accepted: 09/17/2012] [Indexed: 02/06/2023]
Abstract
Invasive bacterial pathogens often target cellular proteins involved in adhesion as a first event during infection. For example, Listeria monocytogenes uses the bacterial protein InlA to interact with E-cadherin, hijack the host adherens junction (AJ) machinery and invade non-phagocytic cells by a clathrin-dependent mechanism. Here, we investigate a potential role for clathrin in cell-cell adhesion. We observed that the initial steps of AJ formation trigger the phosphorylation of clathrin, and its transient localization at forming cell-cell contacts. Furthermore, we show that clathrin serves as a hub for the recruitment of proteins that are necessary for the actin rearrangements that accompany the maturation of AJs. Using an InlA/E-cadherin chimera, we show that adherent cells expressing the chimera form AJs with cells expressing E-cadherin. We demonstrate that non-adherent cells expressing the InlA chimera, as bacteria, can be internalized by E-cadherin-expressing adherent cells. Together these results reveal that a common clathrin-mediated machinery may regulate internalization and cell adhesion and that the relative mobility of one of the interacting partners plays an important role in the commitment to either one of these processes.
Collapse
Affiliation(s)
- Matteo Bonazzi
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris, F-75015, France.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Fletcher NF, Howard C, McKeating JA. Over the fence or through the gate: how viruses infect polarized cells. Immunotherapy 2012; 4:249-51. [PMID: 22401628 DOI: 10.2217/imt.12.6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|