1
|
Merk D, Cox FF, Jakobs P, Prömel S, Altschmied J, Haendeler J. Dose-Dependent Effects of Lipopolysaccharide on the Endothelium-Sepsis versus Metabolic Endotoxemia-Induced Cellular Senescence. Antioxidants (Basel) 2024; 13:443. [PMID: 38671891 PMCID: PMC11047739 DOI: 10.3390/antiox13040443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The endothelium, the innermost cell layer of blood vessels, is not only a physical barrier between the bloodstream and the surrounding tissues but has also essential functions in vascular homeostasis. Therefore, it is not surprising that endothelial dysfunction is associated with most cardiovascular diseases. The functionality of the endothelium is compromised by endotoxemia, the presence of bacterial endotoxins in the bloodstream with the main endotoxin lipopolysaccharide (LPS). Therefore, this review will focus on the effects of LPS on the endothelium. Depending on the LPS concentration, the outcomes are either sepsis or, at lower concentrations, so-called low-dose or metabolic endotoxemia. Sepsis, a life-threatening condition evoked by hyperactivation of the immune response, includes breakdown of the endothelial barrier resulting in failure of multiple organs. A deeper understanding of the underlying mechanisms in the endothelium might help pave the way to new therapeutic options in sepsis treatment to prevent endothelial leakage and fatal septic shock. Low-dose endotoxemia or metabolic endotoxemia results in chronic inflammation leading to endothelial cell senescence, which entails endothelial dysfunction and thus plays a critical role in cardiovascular diseases. The identification of compounds counteracting senescence induction in endothelial cells might therefore help in delaying the onset or progression of age-related pathologies. Interestingly, two natural plant-derived substances, caffeine and curcumin, have shown potential in preventing endothelial cell senescence.
Collapse
Affiliation(s)
- Dennis Merk
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
| | - Fiona Frederike Cox
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
- Medical Faculty, Institute for Translational Pharmacology, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Philipp Jakobs
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
| | - Simone Prömel
- Department of Biology, Institute of Cell Biology, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| | - Joachim Altschmied
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
- Medical Faculty, Cardiovascular Research Institute Düsseldorf, CARID, University Hospital and Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Judith Haendeler
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine-University, 40225 Düsseldorf, Germany; (D.M.); (F.F.C.); (P.J.)
- Medical Faculty, Cardiovascular Research Institute Düsseldorf, CARID, University Hospital and Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
2
|
Chiribao ML, Díaz-Viraqué F, Libisch MG, Batthyány C, Cunha N, De Souza W, Parodi-Talice A, Robello C. Paracrine Signaling Mediated by the Cytosolic Tryparedoxin Peroxidase of Trypanosoma cruzi. Pathogens 2024; 13:67. [PMID: 38251374 PMCID: PMC10818299 DOI: 10.3390/pathogens13010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Peroxiredoxins are abundant and ubiquitous proteins that participate in different cellular functions, such as oxidant detoxification, protein folding, and intracellular signaling. Under different cellular conditions, peroxiredoxins can be secreted by different parasites, promoting the induction of immune responses in hosts. In this work, we demonstrated that the cytosolic tryparedoxin peroxidase of Trypanosoma cruzi (cTXNPx) is secreted by epimastigotes and trypomastigotes associated with extracellular vesicles and also as a vesicle-free protein. By confocal microscopy, we show that cTXNPx can enter host cells by an active mechanism both through vesicles and as a recombinant protein. Transcriptomic analysis revealed that cTXNPx induces endoplasmic reticulum stress and interleukin-8 expression in epithelial cells. This analysis also suggested alterations in cholesterol metabolism in cTXNPx-treated cells, which was confirmed by immunofluorescence showing the accumulation of LDL and the induction of LDL receptors in both epithelial cells and macrophages. BrdU incorporation assays and qPCR showed that cTXNPx has a mitogenic, proliferative, and proinflammatory effect on these cells in a dose-dependent manner. Importantly, we also demonstrated that cTXNPx acts as a paracrine virulence factor, increasing the susceptibility to infection in cTXNPx-pretreated epithelial cells by approximately 40%. Although the results presented in this work are from in vitro studies and likely underestimate the complexity of parasite-host interactions, our work suggests a relevant role for this protein in establishing infection.
Collapse
Affiliation(s)
- María Laura Chiribao
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11000, Uruguay;
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - Florencia Díaz-Viraqué
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - María Gabriela Libisch
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Montevideo 11000, Uruguay;
| | - Narcisa Cunha
- Instituto de Biofísica Carlos Chagas Filho, Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (N.C.); (W.D.S.)
| | - Wanderley De Souza
- Instituto de Biofísica Carlos Chagas Filho, Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (N.C.); (W.D.S.)
| | - Adriana Parodi-Talice
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
- Sección Genética Evolutiva, Instituto de Biología, Facultad de Ciencias, Universidad de la República, Montevideo 11000, Uruguay
| | - Carlos Robello
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11000, Uruguay;
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| |
Collapse
|
3
|
Kotlyarov S. Identification of Important Genes Associated with the Development of Atherosclerosis. Curr Gene Ther 2024; 24:29-45. [PMID: 36999180 DOI: 10.2174/1566523223666230330091241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/06/2022] [Accepted: 01/26/2023] [Indexed: 04/01/2023]
Abstract
Atherosclerosis is one of the most important medical problems due to its prevalence and significant contribution to the structure of temporary and permanent disability and mortality. Atherosclerosis is a complex chain of events occurring in the vascular wall over many years. Disorders of lipid metabolism, inflammation, and impaired hemodynamics are important mechanisms of atherogenesis. A growing body of evidence strengthens the understanding of the role of genetic and epigenetic factors in individual predisposition and development of atherosclerosis and its clinical outcomes. In addition, hemodynamic changes, lipid metabolism abnormalities, and inflammation are closely related and have many overlapping links in regulation. A better study of these mechanisms may improve the quality of diagnosis and management of such patients.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University Named After Academician I.P. Pavlov, Russian Federation
| |
Collapse
|
4
|
Tan K, Dong Y, Tan K, Lim LS, Waiho K, Chen J, Xu P, Kwan KY. siRNA Silencing of FpVtg Induces Ovarian Cell Apoptosis in Redtail Prawn, Fenneropenaeus penicillatus. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1176-1190. [PMID: 38010485 DOI: 10.1007/s10126-023-10269-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023]
Abstract
Inadequate gonadal maturation and poor spawning performance increasingly threaten the sustainability of shrimp aquaculture. Unraveling the mechanisms regulating ovarian development and maturation hence is critical to address industry challenges. Vitellogenin (Vtg), a precursor of yolk protein found in the hepatopancreas and ovary of shrimp, plays a key role in facilitating shrimp's oocyte maturation and embryonic development after oviposition. This study found that FpVtg was specifically expressed in F. penicillatus hepatopancreas and ovary. FpVtg was localized predominantly in the oocyte cytoplasm and distributed uniformly in the hepatopancreas tissue. Silencing FpVtg led to apoptosis in both hepatopancreas and ovary tissues. Furthermore, FpVtg depletion upregulated the expression of ovarian peritrophin 1, ovarian peritrophin 2, serine proteinase inhibitor 6, and juvenile hormone esterase-like carboxylesterase 1, while downregulated that of vitellogenin, delta-9 desaturase, and insulin-like receptor. KEGG pathway analysis implicated such as PI3K-AKT signaling, RNA transport, ECM-receptor interaction, hippo signaling, oocyte meiosis, and apoptosis were enriched and involved in ovarian development. These findings have provided insights into the FpVtg's reproductive role and the associated regulatory genes and pathways in F. penicillatus. This knowledge can contribute to establishing strategies to improve the breeding and aquaculture production of F. penicillatus by elucidating its vitellogenesis regulation in redtail prawn and other penaeid species. Further characterization of the implicated pathways and genes will clarify the intricacies underlying ovarian maturation.
Collapse
Affiliation(s)
- Kianann Tan
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, College of Marine Sciences, Beibu Gulf University, Qinzhou City, 535011, Guangxi, China
| | - Yaxin Dong
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, College of Marine Sciences, Beibu Gulf University, Qinzhou City, 535011, Guangxi, China
| | - Karsoon Tan
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, College of Marine Sciences, Beibu Gulf University, Qinzhou City, 535011, Guangxi, China
| | - Leong-Seng Lim
- Borneo Marine Research Institute, Universiti Malaysia Sabah, Kota Kinabalu City, Sabah, Malaysia
| | - Khor Waiho
- Higher Institution Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, Universiti Malaysia Terengganu, Kuala Nerus City, 21030, Terengganu, Malaysia
| | - Jing Chen
- Zhejiang Institute of Freshwater Fisheries, Huzhou City, 313001, China
| | - Peng Xu
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, College of Marine Sciences, Beibu Gulf University, Qinzhou City, 535011, Guangxi, China.
| | - Kit Yue Kwan
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, College of Marine Sciences, Beibu Gulf University, Qinzhou City, 535011, Guangxi, China.
| |
Collapse
|
5
|
Turman BJ, Darville T, O'Connell CM. Plasmid-mediated virulence in Chlamydia. Front Cell Infect Microbiol 2023; 13:1251135. [PMID: 37662000 PMCID: PMC10469868 DOI: 10.3389/fcimb.2023.1251135] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Chlamydia trachomatis infection of ocular conjunctiva can lead to blindness, while infection of the female genital tract can lead to chronic pelvic pain, ectopic pregnancy, and/or infertility. Conjunctival and fallopian tube inflammation and the resulting disease sequelae are attributed to immune responses induced by chlamydial infection at these mucosal sites. The conserved chlamydial plasmid has been implicated in enhancing infection, via improved host cell entry and exit, and accelerating innate inflammatory responses that lead to tissue damage. The chlamydial plasmid encodes eight open reading frames, three of which have been associated with virulence: a secreted protein, Pgp3, and putative transcriptional regulators, Pgp4 and Pgp5. Although Pgp3 is an important plasmid-encoded virulence factor, recent studies suggest that chlamydial plasmid-mediated virulence extends beyond the expression of Pgp3. In this review, we discuss studies of genital, ocular, and gastrointestinal infection with C. trachomatis or C. muridarum that shed light on the role of the plasmid in disease development, and the potential for tissue and species-specific differences in plasmid-mediated pathogenesis. We also review evidence that plasmid-associated inflammation can be independent of bacterial burden. The functions of each of the plasmid-encoded proteins and potential molecular mechanisms for their role(s) in chlamydial virulence are discussed. Although the understanding of plasmid-associated virulence has expanded within the last decade, many questions related to how and to what extent the plasmid influences chlamydial infectivity and inflammation remain unknown, particularly with respect to human infections. Elucidating the answers to these questions could improve our understanding of how chlamydia augment infection and inflammation to cause disease.
Collapse
Affiliation(s)
- Breanna J. Turman
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Toni Darville
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, United States
| | | |
Collapse
|
6
|
Xiao J, He J, He Z, Wang C, Li Y, Yan X, Chen Y, Sun Z, Liu J, Liang M, Wu Y. Chlamydia psittaci hypothetical inclusion membrane protein CPSIT_0842 evokes a pro-inflammatory response in monocytes via TLR2/TLR4 signaling pathways. Vet Microbiol 2023; 280:109693. [PMID: 36889151 DOI: 10.1016/j.vetmic.2023.109693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 02/05/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023]
Abstract
Chlamydia psittaci (C. psittaci) is an obligate intracellular pathogen that resides within a membrane-bound compartment known as the inclusion. Upon entering the host cell, Chlamydiae secrete numerous proteins to modify the inclusion membrane. Inclusion membrane (Inc) proteins are important pathogenic factors in Chlamydia and play crucial roles in the growth and development of Chlamydia. In the present study, the C. psittaci protein, CPSIT_0842, was identified and shown to localize to the inclusion membrane. Temporal analysis revealed that CPSIT_0842 is an early expression protein of Chlamydia. Moreover, this protein was shown to induce the expression of pro-inflammatory cytokines IL-6 and IL-8 in human monocytes (THP-1 cells) via the TLR2/TLR4 signaling pathway. CPSIT_0842 increases the expression of TLR2, TLR4, and adaptor MyD88. Suppression of TLR2, TLR4, and MyD88 markedly attenuated CPSIT_0842-induced production of IL-6 and IL-8. MAP kinases and NF-κB, important downstream molecules of TLR receptors in inflammatory signaling pathways, were also confirmed to be activated by CPSIT_0842. CPSIT_0842-induced production of IL-6 was reliant on activation of the ERK, p38, and NF-κB signaling pathways while IL-8 expression was regulated by the ERK, JNK, and NF-κB signaling pathways. Specific inhibitors of these signaling pathways significantly decreased CPSIT_0842-mediated expression of IL-6 and IL-8. Together these findings demonstrate that CPSIT_0842 upregulates the expression of IL-6 and IL-8 via TLR-2/TLR4-mediated MAPK and NF-κB signaling pathways in THP-1 cells. Exploring these molecular mechanisms enhances our understanding of C. psittaci pathogenesis.
Collapse
Affiliation(s)
- Jian Xiao
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, 421001, China; The Affiliated Nanhua Hospital, Department of laboratory medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, 421001, China
| | - Jun He
- The Affiliated Nanhua Hospital, Department of laboratory medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhangping He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, 421001, China; The Affiliated Nanhua Hospital, Department of laboratory medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, 421001, China
| | - Chuan Wang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, 421001, China
| | - Yumeng Li
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, 421001, China
| | - Xiaoliang Yan
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, 421001, China
| | - Yuqing Chen
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, 421001, China
| | - Zhenjie Sun
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, 421001, China
| | - Jian Liu
- The Affiliated Nanhua Hospital, Department of laboratory medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Mingxing Liang
- The Affiliated Huaihua Hospital, Department of laboratory medicine, Huaihua, Hunan, 418000, China
| | - Yimou Wu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, 421001, China.
| |
Collapse
|
7
|
Saha A, Ahmed S. The Link Between Heat Shock Proteins, Renin-Angiotensin System, and the Coagulation Cascade in the Pathogenesis of the Coronavirus-19 Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1409:161-171. [PMID: 35882774 DOI: 10.1007/5584_2022_735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
INTRODUCTION Understanding the pathogenesis of COVID-19 is integral for its successful treatment. METHODS Available literature on the relationship between COVID-19, heat shock proteins (HSP), and the renin-angiotensin-aldosterone (RAAS) system were searched and used to hypothesize how HSP can be targeted in COVID-19. RESULTS During SARS-CoV-2 cellular entry, the ACE-2 receptor is downregulated. This leads to the augmentation of angiotensin-2/AT1 receptor axis along with attenuation of the ACE-2/angiotensin1-7/Mas axis. Heat shock proteins are key stabilizing molecules in various pathways.In the heart and vessels, HSP-90 and HSP-60 can facilitate angiotensin-2-mediated myocardial injury and endothelial cell activation. HSP-60-TLR4/CD14 complex formation stabilizes IκB-kinase (IKK) potentiating NF-κB activation. HSPs in lungs and kidneys have antioxidant, vasodilatory, and anti-inflammatory actions and may be protective against the effects of RAAS. Stress-induced HSP-70 has a role in complement-mediated microvascular injury such as has been demonstrated in COVID-19. SARS-CoV-2 can induce autophagy via Beclin-1 and ER (endoplasmic reticular) stress via BIP. These two can be potential targets in the HSP environment. CONCLUSION Various HSP molecules can modulate the effects of the renin-angiotensin-aldosterone (RAAS) system and thus may have a potential role in the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Aritra Saha
- Department of General Medicine, Assam Medical College and Hospital, Dibrugarh, India
| | - Sakir Ahmed
- Department of Clinical Immunology & Rheumatology, Kalinga Institute of Medical Sciences (KIMS), KIIT University, Bhubaneswar, India.
| |
Collapse
|
8
|
Kaizu X, Ying W, Mei-fang W, Li-ming L. 1,25-dihydroxyvitamin D 3 ameliorates high glucose-mediated proliferation, migration, and MCP-1 secretion of vascular smooth muscle cells by inhibiting MAPK phosphorylation. J Int Med Res 2022. [PMCID: PMC9478726 DOI: 10.1177/03000605221121973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objectives To explore the impacts of 1,25-dihydroxyvitamin D3
(1,25(OH)2D3) on the proliferation,
migration, and monocyte chemoattractant protein-1 (MCP-1)
secretion of vascular smooth muscle cells (VSMCs) in a high
glucose environment and its possible mechanism. Methods We extracted VSMCs from the thoracic aorta of a male Sprague–Dawley
rats before culturing them in a 25-mM glucose-containing medium
in the presence or absence of 1,25(OH)2D3
(10−9 –10−7 M). Cell proliferation
was determined by bromodeoxyuridine incorporation assays.
Subsequently, cell migratory capacity was examined by performing
a transwell assay. An enzyme-linked immunosorbent assay was
conducted to assess MCP-1 levels. Protein levels of matrix
metalloproteinase-9 (MMP-9), mitogen-activated protein kinases
(MAPKs), cyclin D1, and phosphorylated MAPKs were determined by
immunoblotting. Results 1,25(OH)2D3 significantly suppressed the
proliferation, migration, and MCP-1 secretion of VSMCs mediated
by high glucose in a dose-dependent manner, diminished the
enhanced protein expression of MMP-9 and cyclin D1, and
attenuated MAPK phosphorylation. The p38 inhibitor SB203580 and
ERK1/2 inhibitor PD98059 suppressed high glucose-mediated
upregulation of MMP-9 and cyclin D1 protein expression and MCP-1
secretion, respectively. Conclusions 1,25(OH)2D3 ameliorates high glucose-mediated
proliferation, migration, and MCP-1 secretion of VSMCs by
inhibiting MAPK phosphorylation, implying a potential
therapeutic approach using 1,25(OH)2D3 for
diabetic macrovascular complications.
Collapse
Affiliation(s)
- Xu Kaizu
- Department of Cardiology, Affiliated Hospital of Putian University, The Third Clinical Medical College of Fujian Medical University, Putian, China
| | - Wu Ying
- Department of Cardiology, Affiliated Hospital of Putian University, The Third Clinical Medical College of Fujian Medical University, Putian, China
| | - Wu Mei-fang
- Department of Cardiology, Affiliated Hospital of Putian University, The Third Clinical Medical College of Fujian Medical University, Putian, China
| | - Lin Li-ming
- Department of Cardiology, Affiliated Hospital of Putian University, The Third Clinical Medical College of Fujian Medical University, Putian, China
| |
Collapse
|
9
|
Bacterial Infections and Atherosclerosis – A Mini Review. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2022. [DOI: 10.22207/jpam.16.3.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Atherosclerosis is the most challenging subsets of coronary artery disease in humans, in which risk factors emerge from childhood, and its prevalence increases with age. Experimental research demonstrates that infections due to bacteria stimulate atherogenic events. Atherosclerosis has complex pathophysiology that is linked with several bacterial infections by damaging the inner arterial wall and heart muscles directly and indirectly by provoking a systemic pro-inflammation and acute-phase protein. Repeated bacterial infections trigger an inflammatory cascade that triggers immunological responses that negatively impact cardiovascular biomarkers includes triglycerides, high-density lipoprotein, C-reactive protein, heat shock proteins, cytokines, fibrinogen, and leukocyte count. Herein, we intended to share the role of bacterial infection in atherosclerosis and evaluate existing evidence of animal and human trials on the association between bacterial infections and atherosclerosis on update.
Collapse
|
10
|
Prasad SK, Bhat S, Shashank D, C R A, R S, Rachtanapun P, Devegowda D, Santhekadur PK, Sommano SR. Bacteria-Mediated Oncogenesis and the Underlying Molecular Intricacies: What We Know So Far. Front Oncol 2022; 12:836004. [PMID: 35480118 PMCID: PMC9036991 DOI: 10.3389/fonc.2022.836004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/22/2022] [Indexed: 01/10/2023] Open
Abstract
Cancers are known to have multifactorial etiology. Certain bacteria and viruses are proven carcinogens. Lately, there has been in-depth research investigating carcinogenic capabilities of some bacteria. Reports indicate that chronic inflammation and harmful bacterial metabolites to be strong promoters of neoplasticity. Helicobacter pylori-induced gastric adenocarcinoma is the best illustration of the chronic inflammation paradigm of oncogenesis. Chronic inflammation, which produces excessive reactive oxygen species (ROS) is hypothesized to cause cancerous cell proliferation. Other possible bacteria-dependent mechanisms and virulence factors have also been suspected of playing a vital role in the bacteria-induced-cancer(s). Numerous attempts have been made to explore and establish the possible relationship between the two. With the growing concerns on anti-microbial resistance and over-dependence of mankind on antibiotics to treat bacterial infections, it must be deemed critical to understand and identify carcinogenic bacteria, to establish their role in causing cancer.
Collapse
Affiliation(s)
- Shashanka K Prasad
- Department of Biotechnology and Bioinformatics, Faculty of Life Sciences, Jagadguru Sri Shivarathreeshwara (JSS) Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Smitha Bhat
- Department of Biotechnology and Bioinformatics, Faculty of Life Sciences, Jagadguru Sri Shivarathreeshwara (JSS) Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Dharini Shashank
- Department of General Surgery, Adichunchanagiri Institute of Medical Sciences, Mandya, India
| | - Akshatha C R
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Sindhu R
- Department of Microbiology, Faculty of Life Sciences, Jagadguru Sri Shivarathreeshwara (JSS) Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Pornchai Rachtanapun
- School of Agro-Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai, Thailand
- Cluster of Agro Bio-Circular-Green Industry (Agro BCG), Chiang Mai University, Chiang Mai, Thailand
| | - Devananda Devegowda
- Centre of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Prasanna K Santhekadur
- Centre of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Sarana Rose Sommano
- Cluster of Agro Bio-Circular-Green Industry (Agro BCG), Chiang Mai University, Chiang Mai, Thailand
- Department of Plant and Soil Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
11
|
Feng Z, Xu L, Xie Z. Receptors for Respiratory Syncytial Virus Infection and Host Factors Regulating the Life Cycle of Respiratory Syncytial Virus. Front Cell Infect Microbiol 2022; 12:858629. [PMID: 35281439 PMCID: PMC8913501 DOI: 10.3389/fcimb.2022.858629] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 12/02/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of lower respiratory tract infections and responsible for a large proportion of mortality in children and the elderly. There are no licensed vaccines available to date. Prophylaxis and therapeutic RSV-specific antibodies are limited to populations at high risk owing to high cost and uncertain clinical value. Receptors and host factors are two determinants important for virus entry and establishment of infection in vivo. The identification and understanding of viral receptors and host factors can help us to gain insight into the pathogenesis of RSV infection. Herein, we reviewed receptors and host factors that have been reported thus far. RSV could bind to CX3C chemokine receptor 1 and heparan sulfate proteoglycans via the G protein, and to nucleolin, insulin-like growth factor-1 receptor, epidermal growth factor, and intercellular adhesion molecule-1 via the F protein. Seven host restriction factors and 13 host factors essential for RSV infection were reviewed. We characterized the functions and their roles in the life cycle of RSV, trying to provide an update on the information of RSV-related receptors and host factors.
Collapse
Affiliation(s)
- Ziheng Feng
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| | - Lili Xu
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Lili Xu,
| | - Zhengde Xie
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Noreen M, Imran M, Safi SZ, Bashir MA, Gul S, Alkhuriji AF, Aloma SY, Alharbi HM, Arshad M. Protective role of TIRAP functional variant against development of coronary artery disease. Saudi J Biol Sci 2021; 28:3548-3552. [PMID: 34121897 PMCID: PMC8176052 DOI: 10.1016/j.sjbs.2021.03.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 11/17/2022] Open
Abstract
Coronary artery disease (CAD) is the leading cause of sudden death worldwide. Inflammation is proved to be an important player in development of the CAD. Inflammation is directly regulated by the Toll like receptors (TLRs). Susceptibility of CAD is influenced by genetic variations within TLRs and the proteins involved in its signaling cascade. The TIRAP/MAL {TIR domain containing adaptor protein / MyD88 (myeloid differentiation primary response gene 88) adaptor-like} exhibits maximum genetic variations of all adaptor proteins involved in TLR signaling cascade. Susceptibility to a number of diseases can be influenced due to presence of S180L single nucleotide polymorphism (SNP) of TIRAP/MAL. This study was conducted to investigate the functional role of this well characterized S180L polymorphism on susceptibility to CAD among Pakistani patients. A total of 146 Pakistani CAD patients and 147 controls were genotyped by Amplification Refractory Mutation System-Polymerase Chain Reaction (ARMS-PCR) and the data was analyzed by using 2-tailed Chi square (x2 ) test. The p value ≤ 0.05 was considered to be significant. Significantly high frequency of homozygous L180L genotype was observed among healthy subjects as compared to the CAD patients [24 (16%) vs 7 (5%); x2 11.85; p = 0.003]. Moreover, the allele frequency of the minor allele; 180L was observed to be significantly higher among controls than the CAD patients, having same direction of association [156 (53%) vs 131 (45%); OR (95% CI) = 0.7198 (0.520-0.996); p < 0.05). Our results indicate that protective effect of L180L; a coding variant of TIRAP/MAL against CAD is discernible.
Collapse
Affiliation(s)
- Mamoona Noreen
- Atta ur Rehman School of Applied Biosciences (ASAB), National University of Sciences & Technology (NUST), H-12 sector, Islamabad, Pakistan
- Department of Zoology, The Women University Multan, Multan, Pakistan
| | - Muhammad Imran
- Department of Microbiology, University of Health Sciences, Lahore, Pakistan
| | - Sher Zaman Safi
- Interdisciplinary Research Center in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology, Lahore, Pakistan
| | - Muhammad Amjad Bashir
- Department of Plant Protection Faculty of Agricultural Sciences, Ghazi University, Dera Ghazi Khan, Pakistan
| | - Sana Gul
- Department of Biological Sciences National University of Medical Sciences (NUMS) Rawalpindi, Pakistan
| | - Afrah Fahad Alkhuriji
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Suliman Yousef Aloma
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Hanan Mualla Alharbi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Arshad
- Atta ur Rehman School of Applied Biosciences (ASAB), National University of Sciences & Technology (NUST), H-12 sector, Islamabad, Pakistan
- University of Education, Lahore, Pakistan
| |
Collapse
|
13
|
Lin HH, Lin YW, Wu CY, Hsiao FP, Lai YL, Hung SL. Stimulatory effects of Porphyromonas gingivalis GroEL protein on interleukin-6 and interleukin-8 in human osteoblasts. J Formos Med Assoc 2021; 120:150-156. [DOI: 10.1016/j.jfma.2020.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 01/08/2023] Open
|
14
|
Hijacking and Use of Host Kinases by Chlamydiae. Pathogens 2020; 9:pathogens9121034. [PMID: 33321710 PMCID: PMC7763869 DOI: 10.3390/pathogens9121034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Chlamydia species are causative agents of sexually transmitted infections, blinding trachoma, and animal infections with zoonotic potential. Being an obligate intracellular pathogen, Chlamydia relies on the host cell for its survival and development, subverting various host cell processes throughout the infection cycle. A key subset of host proteins utilized by Chlamydia include an assortment of host kinase signaling networks which are vital for many chlamydial processes including entry, nutrient acquisition, and suppression of host cell apoptosis. In this review, we summarize the recent advancements in our understanding of host kinase subversion by Chlamydia.
Collapse
|
15
|
Keyoumu Y, Huo Q, Cheng L, Ma H, Zhang M, Ma Y, Ma X. The detailed biological investigations about combined effects of novel polyphenolic and photo-plasmonic nanoparticles loaded graphene nanosheets on coronary endothelial cells and isolated rat aortic rings. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 202:111666. [PMID: 31837585 DOI: 10.1016/j.jphotobiol.2019.111666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/15/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022]
Abstract
In this study, the effect of Polyp-Au-GO nanocomposite on VSMC proliferation, cell cycle proteins, down-regulation of mRNA in the rat was tested. Briefly, Polyp-Au-GO composite material was synthesized and characterized by UV-Vis spectra, X-ray diffraction (XRD), Raman spectroscopy, Fourier transform infrared spectroscopy (FT-IR), Scanning electron microscopy (SEM) and Transmission electron microscopy (TEM). Polyp-Au-GO composite exhibited the absorbance peak at 530 nm. XRD analysis confirmed the crystalline particle with size ranging between 16.5 and 32.6 nm. The crystallinity differences of the nanocomposite were examined by Raman spectroscopy analysis. The presence of a strong band (1500 cm-1) and the absence of other lower frequency bands confirmed that the absence of crystallinity of Polyp-Au-GO nanocomposite. The thermal properties of Polyp-Au-GO nanocomposite were determined by TGA analysis. The results revealed that 15% of its weight loss has occurred at 300 °C. Further, the growth of VSMCs was inhibited by the treatment of Polyp-Au-GO composite at 72 h. The IC50 value was registered at 0.57 μg/mL. Additionally, the Polyp-Au-GO composite arrest G1 cell cycle and down-regulated cell cycle proteins. These Polyp-Au-GO composite also reduced the extracellular ERK1/2 phosphorylation. Furthermore, Polyp-Au-GO composite inhibited TNF-R-evoked inflammatory responses. Moreover, Polyp-Au-GO composite inhibited of CEC proliferation. These results suggest that Polyp-Au-GO composite inhibits VSMC proliferation and TNF-R-mediated inflammatory responses. This study suggested the therapeutic role of Polyp-Au-GO composite in cardiovascular disease.
Collapse
Affiliation(s)
- Yilihamujiang Keyoumu
- Department of cardiovascular surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qiang Huo
- Department of cardiovascular surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Lufeng Cheng
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Hong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xin Jiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Mingming Zhang
- Department of cardiovascular surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yitong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xin Jiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiang Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China; Xin Jiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
16
|
Schnack L, Sohrabi Y, Lagache SMM, Kahles F, Bruemmer D, Waltenberger J, Findeisen HM. Mechanisms of Trained Innate Immunity in oxLDL Primed Human Coronary Smooth Muscle Cells. Front Immunol 2019; 10:13. [PMID: 30728822 PMCID: PMC6351498 DOI: 10.3389/fimmu.2019.00013] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 01/04/2019] [Indexed: 01/29/2023] Open
Abstract
Objective: Damage and pathogen associated molecular patterns such as oxidized low-density lipoprotein (oxLDL) or bacillus Calmette-Guerin (BCG) vaccine can induce long term pro-inflammatory priming in monocytes and macrophages due to metabolic and epigenetic reprogramming—an emerging new concept called trained innate immunity. Vascular smooth muscle cells express pattern recognition receptors involved in trained innate immunity in monocytes. Here we investigated whether the mechanisms of trained innate immunity also control a proinflammatory phenotype in human coronary smooth muscle cells. Methods: Human coronary smooth muscle cells were primed with oxLDL or BCG for 24 h. After a resting time of 4 to 7 days, the cells were restimulated with either PAM3cys4, LPS or TNFα and cytokine production or mRNA expression were measured. Then, mechanisms of monocyte trained innate immunity were analyzed in smooth muscle cells, including receptors, intracellular pathways as well as metabolic and epigenetic reprogramming. Results: Priming with oxLDL or BCG lead to a significantly increased production of IL6, IL8 and MCP-1 following restimulation. OxLDL priming had little effect on the expression of macrophage or SMC marker genes. Proinflammatory priming of smooth muscle cells induced mTOR-HIF1α-signaling and could be blocked by mTOR-, TLR2-, and TLR4-inhibition. Finally, metabolic and epigenetic mechanisms of trained innate immunity in monocytes could be replicated in smooth muscle cells, including increased glucose consumption, lactate production, responsiveness to 6-fluoromevalonate and mevalonate treatment and inhibition of priming by the histone methyltransferase inhibitor methylthioadenosine (MTA). Conclusion: We demonstrate for the first time that mechanisms of the so called trained innate immunity control a proinflammatory phenotype in non-immune cells of the vascular wall. Our findings warrant further research into the specificity of trained innate immunity as an immune cell response as well as the mechanisms of vascular smooth muscle cells inflammation.
Collapse
Affiliation(s)
- Lucia Schnack
- Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| | - Yahya Sohrabi
- Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| | - Sina M M Lagache
- Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| | - Florian Kahles
- Department of Internal Medicine I-Cardiology, University Hospital Aachen, Aachen, Germany
| | - Dennis Bruemmer
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Johannes Waltenberger
- Medical Faculty, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany
| | - Hannes M Findeisen
- Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| |
Collapse
|
17
|
Gomez LM, Anton L, Srinivas SK, Elovitz MA, Parry S. Low-Dose Aspirin May Prevent Trophoblast Dysfunction in Women With Chlamydia Pneumoniae Infection. Reprod Sci 2018; 26:1449-1459. [PMID: 30572799 DOI: 10.1177/1933719118820468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Previously, we demonstrated that live Chlamydia pneumoniae (Cp) impaired extravillous trophoblast (EVT) viability and invasion and that Cp DNA was detected in placentas from cases with preeclampsia. We sought to elucidate whether (1) inactive forms of Cp also affect EVT function; (2) potential therapeutic interventions protect against the effects of Cp; and (3) anti-Cp antibodies are associated with preeclampsia. METHODS Human first-trimester EVTs were infected with ultraviolet light-inactivated Cp. Subgroups of EVTs were pretreated with low-dose acetyl-salicylic acid (ASA), dexamethasone, heparin, and indomethacin. We conducted functional assays after infection with inactivated Cp and measured interleukin 8 (IL8), C-reactive protein (CRP), heat shock protein 60 (HSP60), and tumor necrosis factor-α (TNFα) in culture media. We measured anti-Cp IgG serum levels from women who developed preeclampsia (N = 105) and controls (N = 121). RESULTS Inactivated Cp reduced EVT invasion when compared to noninfected cells (P < .00001) without adversely affecting cell viability. Increased levels of IL8, CRP, HSP60, and TNFα were detected in EVTs infected with inactivated Cp compared to noninfected cells (P < .0001). Only pretreatment with low-dose ASA prevented reduced EVT invasion and decreased release of inflammatory mediators (P < .01). Elevated anti-Cp IgG antibodies were more prevalent in serum from cases with preeclampsia compared to controls (67/105 vs 53/121; adjusted P = .013); elevated IgG correlated significantly with elevated serum CRP and elevated soluble fms-like tyrosine kinase-1-placental growth factor ratio. CONCLUSION Inactivated Cp induces decreased EVT invasion and a proinflammatory response; these effects were abrogated by pretreatment with low-dose ASA. Our results suggest an association between Cp infection, trophoblast dysfunction, and preeclampsia.
Collapse
Affiliation(s)
- Luis M Gomez
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, INOVA Health System, Falls Church, VA, USA
| | - Lauren Anton
- Maternal and Child Health Research Program, University of Pennsylvania, Philadelphia, PA, USA
| | - Shindu K Srinivas
- Maternal and Child Health Research Program, University of Pennsylvania, Philadelphia, PA, USA.,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michal A Elovitz
- Maternal and Child Health Research Program, University of Pennsylvania, Philadelphia, PA, USA.,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel Parry
- Maternal and Child Health Research Program, University of Pennsylvania, Philadelphia, PA, USA.,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
18
|
Huang YH, Lin KH, Yu JS, Wu TJ, Lee WC, Chao CCK, Pan TL, Yeh CT. Targeting HSP60 by subcutaneous injections of jetPEI/HSP60-shRNA destabilizes cytoplasmic survivin and inhibits hepatocellular carcinoma growth. Mol Carcinog 2018; 57:1087-1101. [PMID: 29672920 DOI: 10.1002/mc.22827] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 03/28/2018] [Accepted: 04/17/2018] [Indexed: 01/14/2023]
Abstract
Heat shock protein 60 (HSP60) overexpresses in various types of cancer, but its expression levels and functions in hepatocellular carcinoma (HCC) are still in dispute. We aim to clarify this issue and examine whether HSP60 could be a therapeutic target for HCC. We found drastically enhanced cell apoptosis and suppressed cell proliferation in two HCC cell lines with HSP60-silencing, and also indicated survivin was involved in this regulatory process in vitro and in vivo. However, HSP60-silencing in normal human hepatocytes only resulted in a minimal reduction of cell proliferation but without effects on cell apoptosis. We also showed HSP60 interacted with cytosolic but not mitochondrial survivin by immunoprecipitation assay. A rigorous method was used to standardize quantification from immunoblot assay to obtain more precise expression levels of HSP60 and survivin. The expression of HSP60 and survivin positively correlated in both cancerous and non-cancerous liver tissues (P < 0.001) after analyzing 145 surgically removed HCC tissues. A total of 56.6% of HCC patients overexpressed HSP60 in cancerous tissues, and 40.0% under-expressed HSP60. Higher expression of HSP60 and survivin in non-cancerous tissues both correlated with shorter overall survival (P = 0.029 and P < 0.001, respectively). Finally, we evaluated the therapeutic potential of HSP60 using extraneous delivery of jetPEI/shHSP60 complexes. The treatment results showed significant reduction of tumor weight by 44.3% (P < 0.05), accompanied by under-expression of survivin. These studies suggested that HSP60 not only served as a prognostic marker but also served as a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Ya-Hui Huang
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Kwang-Huei Lin
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Jau-Song Yu
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan, Taiwan.,Molecular Medicine Research Center, Chang-Gung University, Taoyuan, Taiwan
| | - Ting-Jung Wu
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,Division of Liver and Transplantation Surgery, Department of General Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wei-Chen Lee
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,Division of Liver and Transplantation Surgery, Department of General Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chuck C-K Chao
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Tai-Long Pan
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, Chang-Gung University, Taoyuan, Taiwan.,Research Center of Industry of Human Ecology, Chang-Gung University of Science and Technology, Taoyuan, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan, Taiwan.,Molecular Medicine Research Center, Chang-Gung University, Taoyuan, Taiwan
| |
Collapse
|
19
|
Sun D, Wu Y, Wang H, Yan H, Liu W, Yang J. Toll-like receptor 4 rs11536889 is associated with angiographic extent and severity of coronary artery disease in a Chinese population. Oncotarget 2018; 8:2025-2033. [PMID: 28002812 PMCID: PMC5356775 DOI: 10.18632/oncotarget.14014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/07/2016] [Indexed: 02/05/2023] Open
Abstract
Toll-like receptor 4 (TLR4) is a key modulator in many inflammation-related diseases. Polymorphisms in the TLR4 gene may alter TLR4 expression and affect the extent and severity of coronary artery disease (CAD). We analyzed 3 polymorphisms of TLR4 in 607 Chinese subjects who underwent coronary arteriography. Blood samples were collected to identify the polymorphisms. We evaluated the relationships between the polymorphisms and the number of vessels involved in coronary stenosis, Gensini scores, and Duke prognostic scores. We found that rs11536889 was associated with an increased risk of 3-vessel disease. When subjects with 3-vessel disease were compared to subjects with nonsignificant CAD, rs11536889 variant genotypes were associated with an increased risk of 3-vessel disease (GC/CC vs. GG: OR=2.06, 95%CI=1.21-3.51). When subjects with 3-vessel disease were compared to subjects with 1-vessel disease, rs11536889 variant genotypes were associated with an increased risk of 3-vessel disease (GC vs. GG: OR=2.14, 95%CI=1.20-3.79; GC/CC vs. GG: OR=2.06, 95%CI=1.20-3.54). When subjects with 3-vessel disease were compared to subjects with non-3-vessel disease, rs11536889 variant genotypes were associated with an increased risk of 3-vessel disease (GC vs. GG: OR=1.76, 95%CI=1.12-2.75; GC/CC vs. GG: OR=1.83, 95%CI=1.19-2.82). The TLR4 rs11536889 polymorphism was also related to Gensini score (P=0.02). The Gensini score was higher in subjects with the variant CC and GC/CC genotype than in subjects with the wild GG genotype (61.28 1.84 and 57.6434.82 vs. 51.2734.57). Our results demonstrate that TLR4 rs11536889 polymorphism is a novel genetic factor in the development of CAD, influencing the extent and severity of CAD.
Collapse
Affiliation(s)
- Dandan Sun
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yupeng Wu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Honghu Wang
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hong Yan
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wen Liu
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jun Yang
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Liu J, Miao G, Wang B, Zheng N, Ma L, Chen X, Wang G, Zhao X, Zhang L, Zhang L. Chlamydia pneumoniae infection promotes monocyte transendothelial migration by increasing vascular endothelial cell permeability via the tyrosine phosphorylation of VE-cadherin. Biochem Biophys Res Commun 2018; 497:742-748. [PMID: 29462613 DOI: 10.1016/j.bbrc.2018.02.145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 11/28/2022]
Abstract
Migration of monocytes into the subendothelial layer of the intima is one of the critical events in early atherosclerosis. Chlamydia pneumoniae (C. pneumoniae) infection has been shown to promote monocyte transendothelial migration (TEM). However, the exact mechanisms have not yet been fully clarified. In this study, we tested the hypothesis that C. pneumoniae infection increases vascular endothelial cell (VEC) permeability and subsequent monocyte TEM through stimulating the tyrosine phosphorylation of vascular endothelial-cadherin (VE-cadherin). Here, we demonstrated that C. pneumoniae infection promoted monocyte TEM in a TEM assay possibly by increasing the permeability of a VEC line EA.hy926 cell as assessed by measuring the passage of FITC-BSA across a VEC monolayer. Subsequently, Western blot analysis showed that C. pneumoniae infection induced VE-cadherin internalization. Our further data revealed that Src-mediated VE-cadherin phosphorylation at Tyr658 was involved in C. pneumoniae infection-induced internalization of VE-cadherin, VEC hyperpermeability and monocyte TEM. Taken together, our data indicate that C. pneumoniae infection promotes monocyte TEM by increasing VEC permeability via the tyrosine phosphorylation and internalization of VE-cadherin in VECs.
Collapse
Affiliation(s)
- Jingya Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Guolin Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Beibei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Ningbo Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Lu Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Xiaoyu Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Guangyan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Xi Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China.
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China.
| |
Collapse
|
21
|
Gond DP, Singh S, Agrawal NK. Testing an association between TLR4 and CXCR1 gene polymorphisms with susceptibility to urinary tract infection in type 2 diabetes in north Indian population. Gene 2017; 641:196-202. [PMID: 29066305 DOI: 10.1016/j.gene.2017.10.060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Genetic variations of Toll like receptor 4 (TLR4) and CXC-chemokine receptor type1 (CXCR1), the key elements of innate immune system and their association with urinary tract infection (UTI) were studied in general population. In present study we investigate genetic variation of these genes in diabetic patients (3 to 4 times higher prevalence of UTI in comparison to general population). METHODS A total 1100 subjects (318 diabetic patients with UTI, 324 diabetic patients without UTI, 200 non-diabetic UTI patients and 260 age matched healthy control) were enrolled in the study. SNPs of TLR4 rs4986790, rs4986791 and CXCR1 rs2234671 was assessed by PCR-RFLP and PCR-SSP respectively. RESULTS Statistical analysis revealed that A/G genotype and G allele of TLR4 rs4986790 are significantly associated with UTI in both diabetics and nondiabetic patients in comparison to healthy control. Similarly CT genotype and T allele of TLR4 rs4986791 are also significantly associated with UTI in both groups. We also found that prevalence of A/G genotype of TLR4 rs4986790 and CT genotype of TLR4 rs4986791 are significantly higher in patients of diabetes with UTI in comparison to diabetic patients without UTI. We did not find any association of CXCR1 rs2234671 polymorphism with UTI by comparing with any group. CONCLUSION We found that TLR4 rs4986790 and rs4986791 gene polymorphism is a risk for UTI development in both diabetic and nondiabetic patients in north Indian population.
Collapse
Affiliation(s)
- Dinesh Prasad Gond
- Department of Endocrinology and Metabolism, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shivendra Singh
- Department of Nephrology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - N K Agrawal
- Department of Endocrinology and Metabolism, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
22
|
Ciccia F, Rizzo A, Maugeri R, Alessandro R, Croci S, Guggino G, Cavazza A, Raimondo S, Cannizzaro A, Iacopino DG, Salvarani C, Triolo G. Ectopic expression of CXCL13, BAFF, APRIL and LT-β is associated with artery tertiary lymphoid organs in giant cell arteritis. Ann Rheum Dis 2017; 76:235-243. [PMID: 27098405 DOI: 10.1136/annrheumdis-2016-209217] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 12/26/2022]
Abstract
OBJECTIVES To investigate whether artery tertiary lymphoid organs (ATLOs) are present in giant cell arteritis (GCA) and that their formation is associated with the ectopic expression of constitutive lymphoid tissue-homing chemokines. METHODS Reverse transcriptase PCR, immunohistochemical and immunofluorescence analysis were used to determine the presence of ectopic ATLOs in GCA and the expression of chemokines/chemokine receptors and cytokines involved in lymphoneogenesis in the temporal artery samples obtained from 50 patients with GCA and 30 controls. The presence of lymphatic conduits, of follicular dendritic cells (FDCs) precursors and lymphoid tissue inducer cells was also investigated. Finally, expression of CXCL13, B cell activating factor (BAFF), a proliferation-inducing ligand (APRIL) and CCL21 by isolated myofibroblasts was evaluated before and after stimulation with Toll-like receptors (TLRs) agonists and cytokines. RESULTS ATLOs were observed in the media layer of 60% of patients with GCA in close proximity to high endothelial venules and independently by the age of patients and the presence of atherosclerosis. ATLO formation was also accompanied by the expression of CXCL13, BAFF, a proliferation-inducing ligand (APRIL), lymphotoxin (LT)-β, interleukin (IL)-17 and IL-7, the presence of FDC precursors and of lymphoid conduits. Stimulation of myofibroblasts with TLR agonists and cytokines resulted in the upregulation of BAFF and CXCL13. CONCLUSIONS ATLOs occur in the inflamed arteries of patients with GCA possibly representing the immune sites where immune responses towards unknown arterial wall-derived antigens may be organised.
Collapse
Affiliation(s)
- Francesco Ciccia
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
| | - Aroldo Rizzo
- Dipartimento di Oncoematologia, Sezione di Anatomia Patologica, Azienda Ospedaliera Ospedali riuniti Villa Sofia Cervello, Palermo, Italy
| | - Rosario Maugeri
- Dipartimento di Emergenze, Urgenze e Neuroscienze Cliniche, Università di Palermo, Palermo, Italy
| | - Riccardo Alessandro
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Palermo, Italy
| | - Stefania Croci
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Arcispedale Santa Maria Nuova, IRCCS, Reggio Emilia, Italy
| | - Giuliana Guggino
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
| | - Alberto Cavazza
- Pathology Unit, Arcispedale Santa Maria Nuova, IRCCS, Reggio Emilia, Italy
| | - Stefania Raimondo
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Palermo, Italy
| | - Alessandra Cannizzaro
- Dipartimento di Oncoematologia, Sezione di Anatomia Patologica, Azienda Ospedaliera Ospedali riuniti Villa Sofia Cervello, Palermo, Italy
| | | | - Carlo Salvarani
- Unità operativa di Reumatologia, Arcispedale S. Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - Giovanni Triolo
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
| |
Collapse
|
23
|
Knowlton AA. Paying for the Tolls: The High Cost of the Innate Immune System for the Cardiac Myocyte. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1003:17-34. [PMID: 28667552 DOI: 10.1007/978-3-319-57613-8_2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cardiac myocyte differs strikingly from the specialized cells of the immune system, which has two different responses to invading organisms and tissue damage. Adaptive or acquired immunity generates highly specific antibodies in response to threats and is an essential component of immunity; however, adaptive immunity can take 4-7 days to mobilize, and a more primitive response, innate immunity, fills the gap. Innate immunity is expressed in complex and in primitive life forms. Specialized receptors, Toll-like receptors (TLRs), which are widely distributed throughout different tissues recognize danger signals and rapidly respond with the release of noxious substances, such as TNFα. The problem is that many endogenous molecules have been found to act as ligands for specific TLRs, and when these molecules are released into the extracellular environment, they can cause problems by activating innate immunity and an inflammatory response. In cardiac myocytes heat shock protein (HSP)60 can activate TLR4, as can HMGB1, and this type of response can amplify the response to ischemia/reperfusion leading to increased cell and tissue injury. Activation of TLRs can potentially amplify chronic, inflammatory diseases, such as ischemic heart failure. Thus, it is important to understand the regulation of the TLRs and their downstream effects. This chapter will focus on the TLRs and cardiac myocytes.
Collapse
Affiliation(s)
- Anne A Knowlton
- Cardiovascular Division, Department of Medicine, Molecular and Cellular Cardiology, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA. .,Department of Pharmacology, University of California, Davis, CA, USA. .,The Department of Veteran's Affairs, Northern California VA, Sacramento, CA, USA.
| |
Collapse
|
24
|
Hanski L, Vuorela P. Lead Discovery Strategies for Identification of Chlamydia pneumoniae Inhibitors. Microorganisms 2016; 4:E43. [PMID: 27916800 PMCID: PMC5192526 DOI: 10.3390/microorganisms4040043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/28/2016] [Accepted: 11/04/2016] [Indexed: 12/18/2022] Open
Abstract
Throughout its known history, the gram-negative bacterium Chlamydia pneumoniae has remained a challenging target for antibacterial chemotherapy and drug discovery. Owing to its well-known propensity for persistence and recent reports on antimicrobial resistence within closely related species, new approaches for targeting this ubiquitous human pathogen are urgently needed. In this review, we describe the strategies that have been successfully applied for the identification of nonconventional antichlamydial agents, including target-based and ligand-based virtual screening, ethnopharmacological approach and pharmacophore-based design of antimicrobial peptide-mimicking compounds. Among the antichlamydial agents identified via these strategies, most translational work has been carried out with plant phenolics. Thus, currently available data on their properties as antichlamydial agents are described, highlighting their potential mechanisms of action. In this context, the role of mitogen-activated protein kinase activation in the intracellular growth and survival of C. pneumoniae is discussed. Owing to the complex and often complementary pathways applied by C. pneumoniae in the different stages of its life cycle, multitargeted therapy approaches are expected to provide better tools for antichlamydial therapy than agents with a single molecular target.
Collapse
Affiliation(s)
- Leena Hanski
- Pharmaceutical Biology, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.
| | - Pia Vuorela
- Pharmaceutical Biology, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.
| |
Collapse
|
25
|
Cao X, Zhang L, Chen C, Wang Q, Guo L, Ma Q, Deng P, Zhu G, Li B, Pi Y, Long C, Zhang L, Yu Z, Zhou Z, Li J. The critical role of ABCG1 and PPARγ/LXRα signaling in TLR4 mediates inflammatory responses and lipid accumulation in vascular smooth muscle cells. Cell Tissue Res 2016; 368:145-157. [DOI: 10.1007/s00441-016-2518-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/27/2016] [Indexed: 12/15/2022]
|
26
|
Shima K, Coopmeiners J, Graspeuntner S, Dalhoff K, Rupp J. Impact of micro-environmental changes on respiratory tract infections with intracellular bacteria. FEBS Lett 2016; 590:3887-3904. [PMID: 27509029 DOI: 10.1002/1873-3468.12353] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/29/2016] [Accepted: 08/05/2016] [Indexed: 12/26/2022]
Abstract
Community-acquired pneumonia is caused by intra- and extracellular bacteria, with some of these bacteria also being linked to the pathogenesis of chronic lung diseases, including asthma and chronic obstructive pulmonary disease. Chlamydia pneumoniae is an obligate intracellular pathogen that is highly sensitive to micro-environmental conditions controlling both pathogen growth and host immune responses. The availability of nutrients, as well as changes in oxygen, pH and interferon-γ levels, have been shown to directly influence the chlamydial life cycle and clearance. Although the lung has been traditionally regarded as a sterile environment, sequencing approaches have enabled the identification of a large number of bacteria in healthy and diseased lungs. The influence of the lung microbiota on respiratory infections has not been extensively studied so far and data on chlamydial infections are currently unavailable. In the present study, we speculate on how lung microbiota might interfere with acute and chronic infections by focusing exemplarily on the obligate intracellular C. pneumoniae. Furthermore, we consider changes in the gut microbiota as an additional player in the control of lung infections, especially in view the increasing evidence suggesting the involvement of the gut microbiota in various immunological processes throughout the human body.
Collapse
Affiliation(s)
- Kensuke Shima
- Department of Infectious Diseases and Microbiology, University of Lübeck, Germany
| | - Jonas Coopmeiners
- Department of Infectious Diseases and Microbiology, University of Lübeck, Germany
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Germany
| | - Klaus Dalhoff
- Medical Clinic III, University-Hospital Schleswig-Holstein/Campus Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Germany
| |
Collapse
|
27
|
Lee GL, Wu JY, Tsai CS, Lin CY, Tsai YT, Lin CS, Wang YF, Yet SF, Hsu YJ, Kuo CC. TLR4-Activated MAPK-IL-6 Axis Regulates Vascular Smooth Muscle Cell Function. Int J Mol Sci 2016; 17:ijms17091394. [PMID: 27563891 PMCID: PMC5037674 DOI: 10.3390/ijms17091394] [Citation(s) in RCA: 34] [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: 07/01/2016] [Revised: 08/11/2016] [Accepted: 08/17/2016] [Indexed: 11/29/2022] Open
Abstract
Migration of vascular smooth muscle cells (VSMCs) into the intima is considered to be a vital event in the pathophysiology of atherosclerosis. Despite substantial evidence supporting the pathogenic role of Toll-like receptor 4 (TLR4) in the progression of atherogenesis, its function in the regulation of VSMC migration remains unclear. The goal of the present study was to elucidate the mechanism by which TLR4 regulates VSMC migration. Inhibitor experiments revealed that TLR4-induced IL-6 secretion and VSMC migration were mediated via the concerted actions of MyD88 and TRIF on the activation of p38 MAPK and ERK1/2 signaling. Neutralizing anti-IL-6 antibodies abrogated TLR4-driven VSMC migration and F-actin polymerization. Blockade of p38 MAPK or ERK1/2 signaling cascade inhibited TLR4 agonist-mediated activation of cAMP response element binding protein (CREB). Moreover, siRNA-mediated suppression of CREB production repressed TLR4-induced IL-6 production and VSMC migration. Rac-1 inhibitor suppressed TLR4-driven VSMC migration but not IL-6 production. Importantly, the serum level of IL-6 and TLR4 endogenous ligand HMGB1 was significantly higher in patients with coronary artery diseases (CAD) than in healthy subjects. Serum HMGB1 level was positively correlated with serum IL-6 level in CAD patients. The expression of both HMGB1 and IL-6 was clearly detected in the atherosclerotic tissue of the CAD patients. Additionally, there was a positive association between p-CREB and HMGB1 in mouse atherosclerotic tissue. Based on our findings, we concluded that, upon ligand binding, TLR4 activates p38 MAPK and ERK1/2 signaling through MyD88 and TRIF in VSMCs. These signaling pathways subsequently coordinate an additive augmentation of CREB-driven IL-6 production, which in turn triggers Rac-1-mediated actin cytoskeleton to promote VSMC migration.
Collapse
Affiliation(s)
- Guan-Lin Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
- Graduate Institutes of Life Sciences, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Jing-Yiing Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Yi-Ting Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Chin-Sheng Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Yi-Fu Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
- Department of Biochemistry, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
- Graduate Institutes of Life Sciences, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan.
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
28
|
Wu H, Cheng XW, Hu L, Takeshita K, Hu C, Du Q, Li X, Zhu E, Huang Z, Yisireyili M, Zhao G, Piao L, Inoue A, Jiang H, Lei Y, Zhang X, Liu S, Dai Q, Kuzuya M, Shi GP, Murohara T. Cathepsin S Activity Controls Injury-Related Vascular Repair in Mice via the TLR2-Mediated p38MAPK and PI3K-Akt/p-HDAC6 Signaling Pathway. Arterioscler Thromb Vasc Biol 2016; 36:1549-57. [PMID: 27365406 PMCID: PMC4961274 DOI: 10.1161/atvbaha.115.307110] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 06/20/2016] [Indexed: 01/02/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— Cathepsin S (CatS) participates in atherogenesis through several putative mechanisms. The ability of cathepsins to modify histone tail is likely to contribute to stem cell development. Histone deacetylase 6 (HDAC6) is required in modulating the proliferation and migration of various types of cancer cells. Here, we investigated the cross talk between CatS and HADC6 in injury-related vascular repair in mice. Approach and Results— Ligation injury to the carotid artery in mice increased the CatS expression, and CatS-deficient mice showed reduced neointimal formation in injured arteries. CatS deficiency decreased the phosphorylation levels of p38 mitogen-activated protein kinase, Akt, and HDAC6 and toll-like receptor 2 expression in ligated arteries. The genetic or pharmacological inhibition of CatS also alleviated the increased phosphorylation of p38 mitogen-activated protein kinase, Akt, and HDAC6 induced by platelet-derived growth factor BB in cultured vascular smooth muscle cells (VSMCs), and p38 mitogen-activated protein kinase inhibition and Akt inhibition decreased the phospho-HDAC6 levels. Moreover, CatS inhibition caused decrease in the levels of the HDAC6 activity in VSMCs in response to platelet-derived growth factor BB. The HDAC6 inhibitor tubastatin A downregulated platelet-derived growth factor–induced VSMC proliferation and migration, whereas HDAC6 overexpression exerted the opposite effect. Tubastatin A also decreased the intimal VSMC proliferation and neointimal hyperplasia in response to injury. Toll-like receptor 2 silencing decreased the phosphorylation levels of p38 mitogen-activated protein kinase, Akt, and HDAC6 and VSMC migration and proliferation. Conclusions— This is the first report detailing cross-interaction between toll-like receptor 2–mediated CatS and HDAC6 during injury-related vascular repair. These data suggest that CatS/HDAC6 could be a potential therapeutic target for the control of vascular diseases that are involved in neointimal lesion formation.
Collapse
Affiliation(s)
- Hongxian Wu
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Xian Wu Cheng
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.).
| | - Lina Hu
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Kyosuke Takeshita
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Chen Hu
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Qiuna Du
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Xiang Li
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Enbo Zhu
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Zhe Huang
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Maimaiti Yisireyili
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Guangxian Zhao
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Limei Piao
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Aiko Inoue
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Haiying Jiang
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Yanna Lei
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Xiaohong Zhang
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Shaowen Liu
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Qiuyan Dai
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Masafumi Kuzuya
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Guo-Ping Shi
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| | - Toyoaki Murohara
- From the Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.W., Q. Du, S.L., Q. Dai); Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (H.W., X.W.C., K.T., M.Y., T.M.); Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL (C.H., X.Z.); Department of Cardiology, Yanbian University Hospital, Yanji, China (X.W.C., X.L., E.Z., G.Z., L.P., Y.L.); Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan (X.W.C., L.H., A.I., M.K.); Department of Neurology, University of Occupational and Environmental Health, Fukuoka, Japan (Z.H.); Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, China (H.J.); Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea (X.W.C); and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.P.S.)
| |
Collapse
|
29
|
Abstract
Chlamydia pneumoniae, an obligate intracellular bacterial pathogen, has long been investigated as a potential developmental or exacerbating factor in various pathologies. Its unique lifestyle and ability to disseminate throughout the host while persisting in relative safety from the immune response has placed this obligate intracellular pathogen in the crosshairs as a potentially mitigating factor in chronic inflammatory diseases. Many animal model and human correlative studies have been performed to confirm or deny a role for C. pneumoniae infection in these disorders. In some cases, antibiotic clinical trials were conducted to prove a link between bacterial infections and atherosclerosis. In this review, we detail the latest information regarding the potential role that C. pneumoniae infection may have in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Rebecca A Porritt
- Division of Pediatric Infectious Diseases and Immunology, Burns and Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Timothy R Crother
- Division of Pediatric Infectious Diseases and Immunology, Burns and Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
30
|
IRAK1 mediates TLR4-induced ABCA1 downregulation and lipid accumulation in VSMCs. Cell Death Dis 2015; 6:e1949. [PMID: 26512959 PMCID: PMC5399175 DOI: 10.1038/cddis.2015.212] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 06/23/2015] [Accepted: 06/30/2015] [Indexed: 12/14/2022]
Abstract
The activation of Toll-like receptor 4 (TLR4) signaling has an important role in promoting lipid accumulation and pro-inflammatory effects in vascular smooth muscle cells (VSMCs), which facilitate atherosclerosis development and progression. Previous studies have demonstrated that excess lipid accumulation in VSMCs is due to an inhibition of the expression of ATP-binding cassette transporter A1 (ABCA1), an important molecular mediator of lipid efflux from VSMCs. However, the underlying molecular mechanisms of this process are unclear. The purpose of this study was to disclose the underlying molecular mechanisms of TLR4 signaling in regulating ABCA1 expression. Primary cultured VSMCs were stimulated with 50 μg/ml oxidized low-density lipoprotein (oxLDL). We determined that enhancing TLR4 signaling using oxLDL significantly downregulated ABCA1 expression and induced lipid accumulation in VSMCs. However, TLR4 knockout significantly rescued oxLDL-induced ABCA1 downregulation and lipid accumulation. In addition, IL-1R-associated kinase 1 (IRAK1) was involved in the effects of TLR4 signaling on ABCA1 expression and lipid accumulation. Silencing IRAK1 expression using a specific siRNA reversed TLR4-induced ABCA1 downregulation and lipid accumulation in vitro. These results were further confirmed by our in vivo experiments. We determined that enhancing TLR4 signaling by administering a 12-week-long high-fat diet (HFD) to mice significantly increased IRAK1 expression, which downregulated ABCA1 expression and induced lipid accumulation. In addition, TLR4 knockout in vivo reversed the effects of the HFD on IRAK1 and ABCA1 expression, as well as on lipid accumulation. In conclusion, IRAK1 is involved in TLR4-mediated downregulation of ABCA1 expression and lipid accumulation in VSMCs.
Collapse
|
31
|
Zhao Y, Zhang C, Wei X, Li P, Cui Y, Qin Y, Wei X, Jin M, Kohama K, Gao Y. Heat shock protein 60 stimulates the migration of vascular smooth muscle cells via Toll-like receptor 4 and ERK MAPK activation. Sci Rep 2015; 5:15352. [PMID: 26477505 PMCID: PMC4609986 DOI: 10.1038/srep15352] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 09/23/2015] [Indexed: 11/29/2022] Open
Abstract
Accumulating evidence indicates that heat shock protein (HSP) 60 is strongly associated with the pathology of atherosclerosis (AS). However, the precise mechanisms by which HSP60 promotes atherosclerosis remain unclear. In the present study, we found that HSP60 mRNA and protein expression levels in the thoracic aorta are enhanced not only in a mouse model of AS but also in high-fat diet (HFD) mice. HSP60 expression and secretion was activated by platelet-derived growth factor-BB (PDGF-BB) and interleukin (IL)-8 in both human umbilical vein endothelial cells (HUVECs) and vascular smooth muscle cells (VSMCs). HSP60 was found to induce VSMC migration, and exposure to HSP60 activated ERK MAPK signaling. U0126, an inhibitor of ERK, reduced VSMC migration. The HSP60-stimulated VSMCs were found to express TLR4 mRNA but not TLR2 mRNA. Knockdown of TLR4 by siRNA reduced HSP60-induced VSMC migration and HSP60-induced ERK activation. Finally, HSP60 induced IL-8 secretion in VSMCs. Together these results suggest that HSP60 is involved in the stimulation of VSMC migration, via TLR4 and ERK MAPK activation. Meanwhile, activation of HSP60 is one of the most powerful methods of sending a ‘danger signal’ to the immune system to generate IL-8, which assists in the management of an infection or disease.
Collapse
Affiliation(s)
- Ying Zhao
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, 116044, China.,Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Chenxu Zhang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Xuge Wei
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Pei Li
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Ying Cui
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, 116044, China.,Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Yuanhua Qin
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Xiaoqing Wei
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Minli Jin
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, 116044, China
| | - Kazuhiro Kohama
- Research Institute of Pharmaceutical Sciences, Musashino University, Nishitokyo, Tokyo, 2028585, Japan
| | - Ying Gao
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, 116044, China.,Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, China
| |
Collapse
|
32
|
Chlorogenic acid inhibits hypoxia-induced pulmonary artery smooth muscle cells proliferation via c-Src and Shc/Grb2/ERK2 signaling pathway. Eur J Pharmacol 2015; 751:81-8. [DOI: 10.1016/j.ejphar.2015.01.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 01/10/2015] [Accepted: 01/14/2015] [Indexed: 11/20/2022]
|
33
|
Di Pietro M, Filardo S, De Santis F, Mastromarino P, Sessa R. Chlamydia pneumoniae and oxidative stress in cardiovascular disease: state of the art and prevention strategies. Int J Mol Sci 2014; 16:724-35. [PMID: 25561227 PMCID: PMC4307271 DOI: 10.3390/ijms16010724] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/26/2014] [Indexed: 12/15/2022] Open
Abstract
Chlamydia pneumoniae, a pathogenic bacteria responsible for respiratory tract infections, is known as the most implicated infectious agent in atherosclerotic cardiovascular diseases (CVDs). Accumulating evidence suggests that C. pneumoniae-induced oxidative stress may play a critical role in the pathogenesis of CVDs. Indeed, the overproduction of reactive oxygen species (ROS) within macrophages, endothelial cells, platelets and vascular smooth muscle cells (VSMCs) after C. pneumoniae exposure, has been shown to cause low density lipoprotein oxidation, foam cell formation, endothelial dysfunction, platelet adhesion and aggregation, and VSMC proliferation and migration, all responsible for the typical pathological changes of atherosclerotic plaque. The aim of this review is to improve our insight into C. pneumoniae-induced oxidative stress in order to suggest potential strategies for CVD prevention. Several antioxidants, acting on multi-enzymatic targets related to ROS production induced by C. pneumoniae, have been discussed. A future strategy for the prevention of C. pneumoniae-associated CVDs will be to target chlamydial HSP60, involved in oxidative stress.
Collapse
Affiliation(s)
- Marisa Di Pietro
- Department of Public Health and Infectious Diseases, "Sapienza" University, Rome 00185, Italy.
| | - Simone Filardo
- Department of Public Health and Infectious Diseases, "Sapienza" University, Rome 00185, Italy.
| | - Fiorenzo De Santis
- Department of Public Health and Infectious Diseases, "Sapienza" University, Rome 00185, Italy.
| | - Paola Mastromarino
- Department of Public Health and Infectious Diseases, "Sapienza" University, Rome 00185, Italy.
| | - Rosa Sessa
- Department of Public Health and Infectious Diseases, "Sapienza" University, Rome 00185, Italy.
| |
Collapse
|
34
|
Ayyappan JP, Abraham A. Targeting TLR4/MAPKs signaling pathway: A better option for therapeutic inhibition of atherosclerosis. World J Immunol 2014; 4:116-121. [DOI: 10.5411/wji.v4.i2.116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/10/2014] [Accepted: 06/27/2014] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases, especially atherosclerosis, found to be the dreadful diseases worldwide. There are diverse pathways associated with the progression of atherosclerosis. One of the important signaling pathways to target atherosclerotic plaque rupture is toll-like receptor 4 (TLR4) Pathway. Several studies are available for illustrating the role of TLR4 in health and diseases. Different types of immune cell are activated in atherosclerosis but primary cells that are activated by the TLR4 signaling are macrophages and endothelial cells. Mechanisms by which macrophages uptake lipids are diverse and it is very important to target signaling pathway responsible for controlling foam cell formation. The process of macrophages transformed foam cell formation is the critical event in progression of atherosclerotic lesion and TLR4 found to have actively participate in the event through mitogen activated protein kinases (MAPKs) activation. The activation of MAPKs signaling pathway leads to the accumulation of cholesterol in the macrophages and also contribute to the dissociation of IκB and the nuclear translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 subunit, thereby activating key inflammatory cascade activation by MAPKs/NF-κB signaling pathway to induce toxicity by activating different inflammatory parameters. Hence, the review focussed on exploring the role of TLR4/MAPKs signaling pathway for the therapeutic inhibition of atherosclerosis.
Collapse
|
35
|
Eguchi K, Manabe I. Toll-like receptor, lipotoxicity and chronic inflammation: the pathological link between obesity and cardiometabolic disease. J Atheroscler Thromb 2014; 21:629-39. [PMID: 24695021 DOI: 10.5551/jat.22533] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The epidemic growth in the prevalence of obesity has made the impact of metabolic syndrome on cardiovascular events increasingly significant. Elevated visceral adiposity, the indispensable component of metabolic syndrome, is thought to play a primary role in the increasing incidence of cardiometabolic disorders. Importantly, obesity is not merely the simple expansion of adipose tissue mass; it also involves the activation of inflammatory processes within visceral adipose tissue. Adipose tissue inflammation on the one hand enhances the production of proinflammatory adipokines and on the other hand increases the release of free fatty acids via the activation of lipolysis. The adipokines and free fatty acids secreted from visceral fat then contribute to a cardiometabolic pathology. We herein summarize recent advances in our understanding of the mechanisms by which visceral obesity leads to the activation of inflammation in cardiovascular and metabolic tissues and promotes cardiometabolic disease. Our focus is on Toll-like receptor 4 signaling and free fatty acids as mediators of chronic inflammation in patients with metabolic syndrome and atherosclerosis.
Collapse
Affiliation(s)
- Kosei Eguchi
- Department of Cardiovascular Medicine, The University of Tokyo
| | | |
Collapse
|
36
|
Chlamydia pneumoniae antibodies and C-reactive protein levels in patients with abdominal aortic aneurysms. ScientificWorldJournal 2013; 2013:212450. [PMID: 24459421 PMCID: PMC3888745 DOI: 10.1155/2013/212450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 09/19/2013] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION The study aim was to assess the relationship between the presence of antibodies to Chlamydia pneumoniae and abdominal aortic aneurysm (AAA) incidence. PATIENTS AND METHODS Consecutive AAA patients and AAA-free controls were recruited prospectively. Serum samples from both groups were examined to determine Immunoglobulin (Ig) A and IgG titres against Chlamydia pneumoniae by ELISA and C-reactive protein (CRP) concentrations. Results were expressed as mean (SD) or median (IQR) and compared using χ (2) and Mann-Whitney U tests. A P value of <0.05 was considered statistically significant. RESULTS Each study group (AAA/nAAA) comprised 250 patients. 196 (78.7%) AAA patients had positive IgA antichlamydial antibody titres, compared to 181 (72.4%) in the control group (P = 0.008, OR 2.0, 95% CI 1.2-3.5). However, positive IgG antibody titres were similar (191 versus 203; P = 0.222, OR 0.7, 95% CI 0.4-1.3). Average CRP concentrations were higher in AAA individuals. IgA or IgG antibody titres were not related to CRP concentrations. CONCLUSIONS These results demonstrated that the frequent incidence of Chlamydia pneumoniae antibodies within the general population makes it difficult to relate its presence to AAA development, despite the high IgA antibody titres. In addition, raised CRP concentrations in AAA patients are not related to the presence of antichlamydial antibodies.
Collapse
|
37
|
Lin YW, Huang CY, Chen YH, Shih CM, Tsao NW, Lin CY, Chang NC, Tsai CS, Tsai HY, Tsai JC, Huang PH, Li CY, Lin FY. GroEL1, a heat shock protein 60 of Chlamydia pneumoniae, impairs neovascularization by decreasing endothelial progenitor cell function. PLoS One 2013; 8:e84731. [PMID: 24376840 PMCID: PMC3871664 DOI: 10.1371/journal.pone.0084731] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/19/2013] [Indexed: 01/04/2023] Open
Abstract
The number and function of endothelial progenitor cells (EPCs) are sensitive to hyperglycemia, hypertension, and smoking in humans, which are also associated with the development of atherosclerosis. GroEL1 from Chlamydia pneumoniae has been found in atherosclerotic lesions and is related to atherosclerotic pathogenesis. However, the actual effects of GroEL1 on EPC function are unclear. In this study, we investigate the EPC function in GroEL1-administered hind limb-ischemic C57BL/B6 and C57BL/10ScNJ (a toll-like receptor 4 (TLR4) mutation) mice and human EPCs. In mice, laser Doppler imaging, flow cytometry, and immunohistochemistry were used to evaluate the degree of neo-vasculogenesis, circulating level of EPCs, and expression of CD34, vWF, and endothelial nitric oxide synthase (eNOS) in vessels. Blood flow in the ischemic limb was significantly impaired in C57BL/B6 but not C57BL/10ScNJ mice treated with GroEL1. Circulating EPCs were also decreased after GroEL1 administration in C57BL/B6 mice. Additionally, GroEL1 inhibited the expression of CD34 and eNOS in C57BL/B6 ischemic muscle. In vitro, GroEL1 impaired the capacity of differentiation, mobilization, tube formation, and migration of EPCs. GroEL1 increased senescence, which was mediated by caspases, p38 MAPK, and ERK1/2 signaling in EPCs. Furthermore, GroEL1 decreased integrin and E-selectin expression and induced inflammatory responses in EPCs. In conclusion, these findings suggest that TLR4 and impaired NO-related mechanisms could contribute to the reduced number and functional activity of EPCs in the presence of GroEL1 from C. pneumoniae.
Collapse
Affiliation(s)
- Yi-Wen Lin
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chun-Yao Huang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Ming Shih
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Nai-Wen Tsao
- Division of Cardiovascular Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Cheng-Yen Lin
- Department of Computer Science and Information Management, Hung Kuang University, Taichung, Taiwan
| | - Nen-Chung Chang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, National Defense Medical Center, Taipei, Taiwan
| | - Hsiao-Ya Tsai
- Division of Cardiovascular Surgery, National Defense Medical Center, Taipei, Taiwan
| | - Jui-Chi Tsai
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chi-Yuan Li
- Graduate Institute of Clinical Medical Sciences, China Medical University, Taichung, Taiwan
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Feng-Yen Lin
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
38
|
Hu L, Cheng XW, Song H, Inoue A, Jiang H, Li X, Shi GP, Kozawa E, Okumura K, Kuzuya M. Cathepsin K activity controls injury-related vascular repair in mice. Hypertension 2013; 63:607-15. [PMID: 24343118 DOI: 10.1161/hypertensionaha.113.02141] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cathepsin K (CatK) is one of the most potent mammalian collagenases. We showed previously the increased expression of CatK in human and animal atherosclerotic lesions. Here, we hypothesized that ablation of CatK mitigates injury-induced neointimal hyperplasia. Male wild-type (CatK(+/+)) and CatK-deficient (CatK(-/-)) mice underwent ligation or a combination of ligation and polyethylene cuff-replacement injuries to the right common carotid artery just proximal to its bifurcation, and they were then processed for morphological and biochemical studies at specific time points. On operative day 28, CatK(-/-) significantly reduced neointimal formation and neovessel formation in both single- and combination-injured arteries compared with the Cat K(+/+) mice. At early time points, CatK(-/-) reduced the lesion macrophage contents and medial smooth muscle cell proliferation, the mRNA levels of monocyte chemoattractant protein-1, toll-like receptor-2, toll-like receptor-4, chemokine ligand-12, and the gelatinolytic activity related to matrix metalloproteinase-2/-9. An aorta-explant assay revealed that smooth muscle cell movement was impaired in the CatK(-/-) mice compared with the CatK(+/+) mice. In addition, the smooth muscle cells and macrophages from CatK(-/-) mice had less invasive ability through a reconstituted basement membrane barrier. This vasculoprotective effect was mimicked by Cat inhibition with trans-epoxysuccinyl-L-leucylamido-{4-guanidino} butane (E64d). These results demonstrate an essential role of CatK in neointimal lesion formation in response to injury, possibly via the reduction of toll-like receptor-2/-4-mediated inflammation and smooth muscle cell proliferation, suggesting a novel therapeutic strategy for the control of endovascular treatment-related restenosis by regulating CatK activity.
Collapse
Affiliation(s)
- Lina Hu
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan. or
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Di Pietro M, Filardo S, De Santis F, Sessa R. Chlamydia pneumoniae infection in atherosclerotic lesion development through oxidative stress: a brief overview. Int J Mol Sci 2013; 14:15105-20. [PMID: 23877837 PMCID: PMC3742290 DOI: 10.3390/ijms140715105] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/04/2013] [Accepted: 07/10/2013] [Indexed: 12/11/2022] Open
Abstract
Chlamydia pneumoniae, an obligate intracellular pathogen, is known as a leading cause of respiratory tract infections and, in the last two decades, has been widely associated with atherosclerosis by seroepidemiological studies, and direct detection of the microorganism within atheroma. C. pneumoniae is presumed to play a role in atherosclerosis for its ability to disseminate via peripheral blood mononuclear cells, to replicate and persist within vascular cells, and for its pro-inflammatory and angiogenic effects. Once inside the vascular tissue, C. pneumoniae infection has been shown to induce the production of reactive oxygen species in all the cells involved in atherosclerotic process such as macrophages, platelets, endothelial cells, and vascular smooth muscle cells, leading to oxidative stress. The aim of this review is to summarize the data linking C. pneumoniae-induced oxidative stress to atherosclerotic lesion development.
Collapse
Affiliation(s)
- Marisa Di Pietro
- Department of Public Health and Infectious Diseases, “Sapienza” University, Rome 00185, Italy; E-Mails: (M.D.P.); (S.F.); (F.D.S.)
| | - Simone Filardo
- Department of Public Health and Infectious Diseases, “Sapienza” University, Rome 00185, Italy; E-Mails: (M.D.P.); (S.F.); (F.D.S.)
| | - Fiorenzo De Santis
- Department of Public Health and Infectious Diseases, “Sapienza” University, Rome 00185, Italy; E-Mails: (M.D.P.); (S.F.); (F.D.S.)
| | - Rosa Sessa
- Department of Public Health and Infectious Diseases, “Sapienza” University, Rome 00185, Italy; E-Mails: (M.D.P.); (S.F.); (F.D.S.)
| |
Collapse
|
40
|
Parveen N, Varman R, Nair S, Das G, Ghosh S, Mukhopadhyay S. Endocytosis of Mycobacterium tuberculosis heat shock protein 60 is required to induce interleukin-10 production in macrophages. J Biol Chem 2013; 288:24956-71. [PMID: 23846686 DOI: 10.1074/jbc.m113.461004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Understanding the signaling pathways involved in the regulation of anti-inflammatory and pro-inflammatory responses in tuberculosis is extremely important in tailoring a macrophage innate response to promote anti-tuberculosis immunity in the host. Although the role of toll-like receptors (TLRs) in the regulation of anti-inflammatory and pro-inflammatory responses is known, the detailed molecular mechanisms by which the Mycobacterium tuberculosis bacteria modulate these innate responses are not clearly understood. In this study, we demonstrate that M. tuberculosis heat shock protein 60 (Mtbhsp60, Cpn60.1, and Rv3417c) interacts with both TLR2 and TLR4 receptors, but its interaction with TLR2 leads to clathrin-dependent endocytosis resulting in an increased production of interleukin (IL)-10 and activated p38 MAPK. Blockage of TLR2-mediated endocytosis inhibited IL-10 production but induced production of tumor necrosis factor (TNF)-α and activated ERK1/2. In contrast, upon interaction with TLR4, Mtbhsp60 remained predominantly localized on the cell surface due to poorer endocytosis of the protein that led to decreased IL-10 production and p38 MAPK activation. The Escherichia coli homologue of hsp60 was found to be retained mainly on the macrophage surface upon interaction with either TLR2 or TLR4 that triggered predominantly a pro-inflammatory-type immune response. Our data suggest that cellular localization of Mtbhsp60 upon interaction with TLRs dictates the type of polarization in the innate immune responses in macrophages. This information is likely to help us in tailoring the host protective immune responses against M. tuberculosis.
Collapse
Affiliation(s)
- Nazia Parveen
- Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Nampally, Hyderabad 500001 India
| | | | | | | | | | | |
Collapse
|
41
|
Henderson B, Fares MA, Lund PA. Chaperonin 60: a paradoxical, evolutionarily conserved protein family with multiple moonlighting functions. Biol Rev Camb Philos Soc 2013; 88:955-87. [DOI: 10.1111/brv.12037] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 02/20/2013] [Accepted: 03/04/2013] [Indexed: 02/07/2023]
Affiliation(s)
- Brian Henderson
- Department of Microbial Diseases, UCL-Eastman Dental Institute; University College London; London WC1X 8LD U.K
| | - Mario A. Fares
- Department of Genetics; University of Dublin, Trinity College Dublin; Dublin 2 Ireland
- Department of Abiotic Stress; Instituto de Biologia Molecular y Celular de Plantas, Consejo Superior de Investigaciones Cientificas (CSIC-UPV); Valencia 46022 Spain
| | - Peter A. Lund
- School of Biosciences; University of Birmingham; Birmingham B15 2TT U.K
| |
Collapse
|
42
|
Rivera J, Walduck AK, Strugnell RA, Sobey CG, Drummond GR. Chlamydia pneumoniae induces a pro-inflammatory phenotype in murine vascular smooth muscle cells independently of elevating reactive oxygen species. Clin Exp Pharmacol Physiol 2012; 39:218-26. [PMID: 22211630 DOI: 10.1111/j.1440-1681.2011.05657.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
NADPH oxidases (Nox) are reactive oxygen species (ROS)-generating enzymes that play important physiological roles in host defence and redox signalling. However, Nox activity is upregulated in the vascular wall during atherosclerosis and contributes to plaque formation by promoting oxidative stress and inflammation. The bacterium Chlamydia pneumoniae has been detected in vascular smooth muscle cells (VSMC) of human atheroma. We hypothesized that C. pneumoniae infection of VSMC causes Nox activation, which initially limits infection but ultimately causes oxidative stress, activation of pro-inflammatory pathways and an atherogenic phenotype. Chlamydia pneumoniae infection of mouse cultured VSMC significantly increased ROS production by twofold but did not upregulate mRNA expression of Nox1 or Nox4. Chlamydia pneumoniae did increase Nox2 mRNA levels significantly by threefold, but this did not translate to elevated Nox2 protein expression. The Nox inhibitor gp91ds-tat had no effect on C. pneumoniae-induced ROS production. In contrast, apocynin significantly reduced ROS levels by 75% in C. pneumoniae-infected VSMC, an effect most likely attributable to its direct anti-oxidant action. Although apocynin had no effect on C. pneumoniae-induced expression of inflammatory markers, bacteria recovered from apocynin-treated VSMC displayed a higher degree of infectivity in HEp-2 cells. In conclusion, C. pneumoniae infection increases ROS production in VSMC independently of Nox activity. Although elevated ROS production appears to serve a protective role by limiting the spread of infection, we speculate that this response will be detrimental over the long term by causing oxidative stress and a smouldering inflammatory response by maintaining C. pneumoniae persistence within the cell.
Collapse
Affiliation(s)
- Jennifer Rivera
- Department of Pharmacology, Vascular Biology & Immunopharmacology Group, Monash University, Clayton, Australia
| | | | | | | | | |
Collapse
|
43
|
Toll-like receptor 9 activation: a novel mechanism linking placenta-derived mitochondrial DNA and vascular dysfunction in pre-eclampsia. Clin Sci (Lond) 2012; 123:429-35. [PMID: 22671429 DOI: 10.1042/cs20120130] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Emerging evidence suggests that in addition to being the 'power houses' of our cells, mitochondria facilitate effector responses of the immune system. Cell death and injury result in the release of mtDNA (mitochondrial DNA) that acts via TLR9 (Toll-like receptor 9), a pattern recognition receptor of the immune system which detects bacterial and viral DNA but not vertebrate DNA. The ability of mtDNA to activate TLR9 in a similar fashion to bacterial DNA stems from evolutionarily conserved similarities between bacteria and mitochondria. mtDNA may be the trigger of systemic inflammation in pathologies associated with abnormal cell death. PE (pre-eclampsia) is a hypertensive disorder of pregnancy with devastating maternal and fetal consequences. The aetiology of PE is unknown and removal of the placenta is the only effective cure. Placentas from women with PE show exaggerated necrosis of trophoblast cells, and circulating levels of mtDNA are higher in pregnancies with PE. Accordingly, we propose the hypothesis that exaggerated necrosis of trophoblast cells results in the release of mtDNA, which stimulates TLR9 to mount an immune response and to produce systemic maternal inflammation and vascular dysfunction that lead to hypertension and IUGR (intra-uterine growth restriction). The proposed hypothesis implicates mtDNA in the development of PE via activation of the immune system and may have important preventative and therapeutic implications, because circulating mtDNA may be potential markers of early detection of PE, and anti-TLR9 treatments may be promising in the management of the disease.
Collapse
|
44
|
Huang CY, Shih CM, Tsao NW, Chen YH, Li CY, Chang YJ, Chang NC, Ou KL, Lin CY, Lin YW, Nien CH, Lin FY. GroEL1, from Chlamydia pneumoniae, induces vascular adhesion molecule 1 expression by p37(AUF1) in endothelial cells and hypercholesterolemic rabbit. PLoS One 2012; 7:e42808. [PMID: 22900050 PMCID: PMC3416774 DOI: 10.1371/journal.pone.0042808] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 07/11/2012] [Indexed: 12/20/2022] Open
Abstract
The expression of vascular adhesion molecule-1 (VCAM-1) by endothelial cells may play a major role in atherogenesis. The actual mechanisms of chlamydia pneumoniae (C. pneumoniae) relate to atherogenesis are unclear. We investigate the influence of VCAM-1 expression in the GroEL1 from C. pneumoniae-administered human coronary artery endothelial cells (HCAECs) and hypercholesterolemic rabbits. In this study, we constructed the recombinant GroEL1 from C. pneumoniae. The HCAECs/THP-1 adhesion assay, tube formation assay, western blotting, enzyme-linked immunosorbent assay, actinomycin D chase experiment, luciferase reporter assay, and immunohistochemical stainings were performed. The results show that GroEL1 increased both VCAM-1expression and THP-1 cell adhesives, and impaired tube-formation capacity in the HCAECs. GroEL1 significantly increased the VCAM-1 mRNA stability and cytosolic AU-binding factor 1 (AUF1) level. Overexpression of the p37AUF1 significantly increased VCAM-1 gene expression in GroEL1-induced bovine aortic endothelial cells (BAECs). GroEL1 prolonged the stability of VCAM-1 mRNA by increasing both p37AUF1 and the regulation of the 5′ untranslated region (UTR) of the VCAM-1 mRNA in BAECs. In hypercholesterolemic rabbits, GroEL1 administration enhanced fatty-streak and macrophage infiltration in atherosclerotic lesions, which may be mediated by elevated VCAM-1 expression. In conclusion, GroEL1 induces VCAM-1 expression by p37AUF1 in endothelial cells and enhances atherogenesis in hypercholesterolemic rabbits.
Collapse
Affiliation(s)
- Chun-Yao Huang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Research Center For Biomedical Implants and Microsurgery Devices, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chun-Ming Shih
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Nai-Wen Tsao
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Chi-Yuan Li
- Graduate Institute of Clinical Medical Sciences, China Medical University and Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Jia Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Nen-Chung Chang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Keng-Liang Ou
- Research Center For Biomedical Implants and Microsurgery Devices, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Yen Lin
- Department of Computer Science and Information Management, Hung Kuang University, Taichung, Taiwan
| | - Yi-Wen Lin
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chih-Hao Nien
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Feng-Yen Lin
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
45
|
Beckett EL, Phipps S, Starkey MR, Horvat JC, Beagley KW, Foster PS, Hansbro PM. TLR2, but not TLR4, is required for effective host defence against Chlamydia respiratory tract infection in early life. PLoS One 2012; 7:e39460. [PMID: 22724018 PMCID: PMC3378543 DOI: 10.1371/journal.pone.0039460] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 05/21/2012] [Indexed: 12/14/2022] Open
Abstract
Chlamydia pneumoniae commonly causes respiratory tract infections in children, and epidemiological investigations strongly link infection to the pathogenesis of asthma. The immune system in early life is immature and may not respond appropriately to pathogens. Toll-like receptor (TLR)2 and 4 are regarded as the primary pattern recognition receptors that sense bacteria, however their contribution to innate and adaptive immunity in early life remains poorly defined. We investigated the role of TLR2 and 4 in the induction of immune responses to Chlamydia muridarum respiratory infection, in neonatal wild-type (Wt) or TLR2-deficient (−/−), 4−/− or 2/4−/− BALB/c mice. Wt mice had moderate disease and infection. TLR2−/− mice had more severe disease and more intense and prolonged infection compared to other groups. TLR4−/− mice were asymptomatic. TLR2/4−/− mice had severe early disease and persistent infection, which resolved thereafter consistent with the absence of symptoms in TLR4−/− mice. Wt mice mounted robust innate and adaptive responses with an influx of natural killer (NK) cells, neutrophils, myeloid (mDCs) and plasmacytoid (pDCs) dendritic cells, and activated CD4+ and CD8+ T-cells into the lungs. Wt mice also had effective production of interferon (IFN)γ in the lymph nodes and lung, and proliferation of lymph node T-cells. TLR2−/− mice had more intense and persistent innate (particularly neutrophil) and adaptive cell responses and IL-17 expression in the lung, however IFNγ responses and T-cell proliferation were reduced. TLR2/4−/− mice had reduced innate and adaptive responses. Most importantly, neutrophil phagocytosis was impaired in the absence of TLR2. Thus, TLR2 expression, particularly on neutrophils, is required for effective control of Chlamydia respiratory infection in early life. Loss of control of infection leads to enhanced but ineffective TLR4-mediated inflammatory responses that prolong disease symptoms. This indicates that TLR2 agonists may be beneficial in the treatment of early life Chlamydia infections and associated diseases.
Collapse
Affiliation(s)
- Emma L. Beckett
- Centre for Asthma and Respiratory Disease, The University of Newcastle, and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Simon Phipps
- School of Biomedical Sciences and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Malcolm R. Starkey
- Centre for Asthma and Respiratory Disease, The University of Newcastle, and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Jay C. Horvat
- Centre for Asthma and Respiratory Disease, The University of Newcastle, and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Kenneth W. Beagley
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Paul S. Foster
- Centre for Asthma and Respiratory Disease, The University of Newcastle, and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- * E-mail: (PMH); (PSF)
| | - Philip M. Hansbro
- Centre for Asthma and Respiratory Disease, The University of Newcastle, and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- * E-mail: (PMH); (PSF)
| |
Collapse
|
46
|
Wu D, Sheu JS, Liu HC, Yuan RY, Yu JM, Sheu JJ, Hung CH, Hu CJ. Increase of toll-like receptor 4 but decrease of interleukin-8 mRNA expression among ischemic stroke patients under aspirin treatment. Clin Biochem 2012; 45:1316-9. [PMID: 22580394 DOI: 10.1016/j.clinbiochem.2012.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 04/22/2012] [Accepted: 04/23/2012] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Toll-like receptors (TLRs) are molecules conserved in evolution for detecting pathogen invasions and tissue damage and are involved in atherogenesis. This study explores the mRNA expression of TLRs and their probable role in further disease occurrence among ischemic stroke patients. DESIGN AND METHODS A total of 89 ischemic stroke patients and 166 controls were recruited for this study. Total RNA was extracted and mRNA was reverse-transcribed to cDNA and was analyzed for TLRs and interleukin 8 (IL8). RESULTS The TLR4 mRNA expression level is significantly higher in the stroke group. Conversely, IL-8 mRNA levels decreased significantly in the patient group. CONCLUSION Our results suggest that TLR4 overexpression in mRNA levels is observed in stroke patients, which might account for the probable inflammatory injury before or after stroke. A reduction of IL-8 expression could result from the downregulatory effects of aspirin.
Collapse
Affiliation(s)
- Dean Wu
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Chahine MN, Deniset J, Dibrov E, Hirono S, Blackwood DP, Austria JA, Pierce GN. Oxidized LDL promotes the mitogenic actions of Chlamydia pneumoniae in vascular smooth muscle cells. Cardiovasc Res 2012; 92:476-83. [PMID: 22072707 DOI: 10.1093/cvr/cvr251] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS The atherogenic actions of Chlamydia pneumoniae (C. pneumoniae), a common respiratory pathogen, are dependent upon a high-cholesterol environment in vivo. It is possible that oxidized low-density lipoprotein (oxLDL) is responsible for promoting the atherogenic effects of C. pneumoniae through a stimulation of cell proliferation. This study determined whether oxLDL can enhance the mitogenic action of C. pneumoniae in vascular smooth muscle cells (VSMCs) and the involvement of mitogen-activated protein kinase (MAPK) pathways and heat shock protein 60 (HSP60) in these mechanisms. METHODS AND RESULTS Primary rabbit VSMCs were treated with live C. pneumoniae, heat-inactivated C. pneumoniae or infection medium, and subsequently incubated for up to 48 h in the presence or absence of oxLDL. Chlamydia pneumoniae infection alone stimulated cell proliferation and the addition of oxLDL significantly amplified this proliferative effect. This proliferation was accompanied by extracellular signal-regulated kinase-1 and -2 (ERK1/2) activation and an up-regulation of HSP60 expression. Changes in proliferation and HSP60 expression were attenuated by the inhibition of ERK1/2. CONCLUSION These results indicate a novel role for oxLDL in promoting the mitogenic actions of C. pneumoniae in the vasculature. ERK1/2 is an important factor in the stress-mediated response and HSP60 up-regulation in VSMC. These data provide mechanistic evidence that C. pneumoniae may stimulate atherogenesis.
Collapse
Affiliation(s)
- Mirna N Chahine
- Institute of Cardiovascular Sciences, St Boniface Hospital Research Centre, 351 Tache Ave., Winnipeg, Manitoba, Canada R2H 2A6
| | | | | | | | | | | | | |
Collapse
|
48
|
Eitel J, Meixenberger K, van Laak C, Orlovski C, Hocke A, Schmeck B, Hippenstiel S, N'Guessan PD, Suttorp N, Opitz B. Rac1 regulates the NLRP3 inflammasome which mediates IL-1beta production in Chlamydophila pneumoniae infected human mononuclear cells. PLoS One 2012; 7:e30379. [PMID: 22276187 PMCID: PMC3262829 DOI: 10.1371/journal.pone.0030379] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 12/19/2011] [Indexed: 01/19/2023] Open
Abstract
Chlamydophila pneumoniae causes acute respiratory tract infections and has been associated with development of asthma and atherosclerosis. The production of IL-1β, a key mediator of acute and chronic inflammation, is regulated on a transcriptional level and additionally on a posttranslational level by inflammasomes. In the present study we show that C. pneumoniae-infected human mononuclear cells produce IL-1β protein depending on an inflammasome consisting of NLRP3, the adapter protein ASC and caspase-1. We further found that the small GTPase Rac1 is activated in C. pneumoniae-infected cells. Importantly, studies with specific inhibitors as well as siRNA show that Rac1 regulates inflammasome activation in C. pneumoniae-infected cells. In conclusion, C. pneumoniae infection of mononuclear cells stimulates IL-1β production dependent on a NLRP3 inflammasome-mediated processing of proIL-1β which is controlled by Rac1.
Collapse
Affiliation(s)
- Julia Eitel
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Karolin Meixenberger
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia van Laak
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Christine Orlovski
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Hocke
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Schmeck
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philippe Dje N'Guessan
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bastian Opitz
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
49
|
Deniset JF, Hedley TE, Dibrov E, Pierce GN. Chlamydophila pneumoniae infection induces alterations in vascular contractile responses. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1264-1272. [PMID: 22214836 DOI: 10.1016/j.ajpath.2011.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 10/11/2011] [Accepted: 11/11/2011] [Indexed: 11/29/2022]
Abstract
Chlamydophila pneumoniae infection has been associated in previous studies with coronary artery disease. The live bacterium has been detected within atherosclerotic plaques and can induce the structural remodeling of the vessel wall. However, the direct effects of infection on the contractile characteristics of the arteries remain unknown. Left anterior descending coronary arteries isolated from porcine hearts were dissected and placed in culture medium for 72 hours before infection with C. pneumoniae. Contractile responses to high molar KCl and u46619 levels and relaxation responses to bradykinin and sodium nitroprusside were assessed at days 5 and 10 postinfection. C. pneumoniae induced decreases in both KCl- and u46619-induced contractile responses at both time points. The altered contractile responses coincided with a down-regulation of L-type Ca(2+) channels at both time points and inositol 1,4,5-triphosphate receptor (IP3R) levels at day 10 postinfection. Infection also induced attenuation of the endothelial-dependent relaxation response to bradykinin at day 10 postinfection. A decrease in endothelial nitric oxide synthase expression levels was noted at day 10 postinfection. Furthermore, an increase in superoxide production combined with an increase in p22phox expression levels was also observed at this time point. These findings indicate that C. pneumoniae infection can directly alter the vascular contractile responses in porcine coronary arteries, providing additional evidence for the role of C. pneumoniae infection in cardiovascular disease.
Collapse
Affiliation(s)
- Justin F Deniset
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, Department of Physiology, Faculties of Medicine and Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Thomas E Hedley
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, Department of Physiology, Faculties of Medicine and Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Elena Dibrov
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, Department of Physiology, Faculties of Medicine and Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Grant N Pierce
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, Department of Physiology, Faculties of Medicine and Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
50
|
Abstract
Bartonella spp. are facultative intracellular pathogens that employ a unique stealth infection strategy comprising immune evasion and modulation, intimate interaction with nucleated cells, and intraerythrocytic persistence. Infections with Bartonella are ubiquitous among mammals, and many species can infect humans either as their natural host or incidentally as zoonotic pathogens. Upon inoculation into a naive host, the bartonellae first colonize a primary niche that is widely accepted to involve the manipulation of nucleated host cells, e.g., in the microvasculature. Consistently, in vitro research showed that Bartonella harbors an ample arsenal of virulence factors to modulate the response of such cells, gain entrance, and establish an intracellular niche. Subsequently, the bacteria are seeded into the bloodstream where they invade erythrocytes and give rise to a typically asymptomatic intraerythrocytic bacteremia. While this course of infection is characteristic for natural hosts, zoonotic infections or the infection of immunocompromised patients may alter the path of Bartonella and result in considerable morbidity. In this review we compile current knowledge on the molecular processes underlying both the infection strategy and pathogenesis of Bartonella and discuss their connection to the clinical presentation of human patients, which ranges from minor complaints to life-threatening disease.
Collapse
Affiliation(s)
- Alexander Harms
- Focal Area Infection Biology, Biozentrum, University of Basel, Switzerland
| | | |
Collapse
|