1
|
Bio-active components in medicinal plants: A mechanistic review of their effects on fish growth and physiological parameters. ANNALS OF ANIMAL SCIENCE 2022. [DOI: 10.2478/aoas-2022-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
World population is increasing at a tremendous rate so is the demand for animal-based protein. Aquaculture is a promising industry that has the potential to supply high quality protein for mankind with minimum environmental impact. In the past decade, aquaculture practices have been shifting from extensive to intensive culture. To achieve maximum production per unit area, high stocking densities are maintained in intensive aquaculture. If not managed properly, this may lead to stress in fish. Fish under stress condition show decreased growth, suppressed appetite, weakened immunity and increased susceptibility to infections. Chemicals, vaccines and antibiotics are used for the treatment of diseased fish. Use of synthetic chemicals, vaccines and antibiotics is not sustainable because pathogens develop resistance against them and they have high residues. Moreover, certain chemicals used for the treatment of fish diseases are not safe for humans therefore, are banned in some countries. Plant parts and their extracts are used in traditional medicines to cure many diseases and to improve health of mankind. In aquaculture industry, use of plants and their derivatives in fish feed to improve health status of fish is increasing. Several plants improve growth and overall health status of fish, some provide protection against pathogens by improving the immune system while others increase appetite by direct action on neuro-endocrine axis of fish. This review provides an in depth and up to date information about use of medicinal plants and their derivatives to improve growth and physiological status of fish and their possible mechanism of action.
Collapse
|
2
|
Wang L, Xie X, Ke B, Huang W, Jiang X, He G. Recent advances on endogenous gasotransmitters in inflammatory dermatological disorders. J Adv Res 2021; 38:261-274. [PMID: 35572410 PMCID: PMC9091779 DOI: 10.1016/j.jare.2021.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/20/2022] Open
Abstract
Endogenous gasotransmitters nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), and potential candidates sulfur dioxide (SO2), methane (CH4), hydrogen gas (H2), ammonia (NH3) and carbon dioxide (CO2), are generated within the human body. Endogenous and potential gasotransmitters regulate inflammation, vasodilation, and oxidation in inflammatory dermatological disorders. Endogenous and potential gasotransmitters play potential roles in psoriasis, atopic dermatitis, acne, and chronic skin ulcers. Further research should explore the function of these gases and gas donors and inhibitors in inflammatory dermatological disorders.
Background Endogenous gasotransmitters are small gaseous mediators that can be generated endogenously by mammalian organisms. The dysregulation of the gasotransmitter system is associated with numerous disorders ranging from inflammatory diseases to cancers. However, the relevance of these endogenous gasotransmitters, prodrug donors and inhibitors in inflammatory dermatological disorders has not yet been thoroughly reviewed and discussed. Aim of review This review discusses the recent progress and will provide perspectives on endogenous gasotransmitters in the context of inflammatory dermatological disorders. Key scientific concepts of review Endogenous gasotransmitters nitric oxide (NO), carbon monoxide (CO) and hydrogen sulfide (H2S) are signaling molecules that regulate several physiological and pathological processes. In addition, sulfur dioxide (SO₂), methane (CH4), hydrogen gas (H2), ammonia (NH3), and carbon dioxide (CO2) can also be generated endogenously and may take part in physiological and pathological processes. These signaling molecules regulate inflammation, vasodilation, and oxidative stress, offering therapeutic potential and attracting interest in the field of inflammatory dermatological disorders including psoriasis, atopic dermatitis, acne, rosacea, and chronic skin ulcers. The development of effective gas donors and inhibitors is a promising alternative to treat inflammatory dermatological disorders with controllable and precise delivery in the future.
Collapse
Affiliation(s)
- Lian Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Xin Xie
- College of Medical Technology and School of Pharmacy, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Bowen Ke
- Laboratory of Anaesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Corresponding authors at: Department of Dermatology, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu 610041, China (X. Jiang and G. He). Laboratory of Anaesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu 610041, China (B.-W. Ke).
| | - Wei Huang
- College of Medical Technology and School of Pharmacy, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
- Corresponding authors at: Department of Dermatology, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu 610041, China (X. Jiang and G. He). Laboratory of Anaesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu 610041, China (B.-W. Ke).
| | - Gu He
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
- Corresponding authors at: Department of Dermatology, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu 610041, China (X. Jiang and G. He). Laboratory of Anaesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Wuhou District, Chengdu 610041, China (B.-W. Ke).
| |
Collapse
|
3
|
Anti-Inflammatory Effects of Abeliophyllum distichum Nakai (Cultivar Okhwang 1) Callus through Inhibition of PI3K/Akt, NF-κB, and MAPK Signaling Pathways in Lipopolysaccharide-Induced Macrophages. Processes (Basel) 2021. [DOI: 10.3390/pr9061071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
One of the Korean endemic plants, Abeliophyllum distichum Nakai (Oleaceae), contains acteoside, which is a glycoside exhibiting neuroprotective, anti-inflammation effects and antibacterial capacities. We conducted an investigation on the effects of the callus of A. distichum (cultivar Okhwang 1, CAO) on pro-inflammatory mediators released following nuclear factor-кB (NF-кB), phosphatidylinositol 3-kinase/Akt (PI3K-Akt) and mitogen-activated protein kinase (MAPK) signal activation in lipopolysaccharide (LPS)-induced RAW 264.7 cells. Immunoblotting was employed to find out the expression of cyclooxygenase-2 (COX-2), inducible nitric oxide (iNOS), and activation of MAPK molecules, NF-κB and Akt. Cytokines, COX-2, and iNOS gene expression were assessed using polymerase chain reaction techniques. Cytokines, COX-2, and iNOS gene expression were assessed using polymerase chain reaction techniques. High-performance liquid chromatography revealed that CAO was rich in acteoside and isoacteoside. As a result, CAO inhibited the generation of NO, cytokines, COX-2, and iNOS expression. Further, translocation to the nuclear of NF-κB p65 and degradation of the inhibitor of NF-кB (IкB) were alleviated by suppressing phosphorylation. Additionally, CAO significantly impacted MAPK pathway activation by potentially reducing phosphorylation of MAPKs. These results indicate that the anti-inflammatory effect of CAO is mediated via the inhibition of MAPK, PI3K/Akt, and NF-κB signaling pathways, probably via glycosides, phenolics, and flavonoids bioactivity derived from plants. CAO can serve as a potential anti-inflammatory agent, which alleviates inflammation factors and act through specific cell signaling pathways.
Collapse
|
4
|
Valente Duarte De Sousa IC. New and emerging drugs for the treatment of acne vulgaris in adolescents. Expert Opin Pharmacother 2019; 20:1009-1024. [DOI: 10.1080/14656566.2019.1584182] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
5
|
Liu C, Liang MC, Soong TW. Nitric Oxide, Iron and Neurodegeneration. Front Neurosci 2019; 13:114. [PMID: 30833886 PMCID: PMC6388708 DOI: 10.3389/fnins.2019.00114] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/30/2019] [Indexed: 12/25/2022] Open
Abstract
Iron is a crucial cofactor for several physiological functions in the brain including transport of oxygen, DNA synthesis, mitochondrial respiration, synthesis of myelin, and neurotransmitter metabolism. If iron concentration exceeds the capacity of cellular sequestration, excessive labile iron will be harmful by generating oxidative stress that leads to cell death. In patients suffering from Parkinson disease, the total amount of iron in the substantia nigra was reported to increase with disease severity. High concentrations of iron were also found in the amyloid plaques and neurofibrillary tangles of human Alzheimer disease brains. Besides iron, nitric oxide (NO) produced in high concentration has been associated with neurodegeneration. NO is produced as a co-product when the enzyme NO synthase converts L-arginine to citrulline, and NO has a role to support normal physiological functions. When NO is produced in a high concentration under pathological conditions such as inflammation, aberrantly S-nitrosylated proteins can initiate neurodegeneration. Interestingly, NO is closely related with iron homeostasis. Firstly, it regulates iron-related gene expression through a system involving iron regulatory protein and its cognate iron responsive element (IRP-IRE). Secondly, it modified the function of iron-related protein directly via S-nitrosylation. In this review, we examine the recent advances about the potential role of dysregulated iron homeostasis in neurodegeneration, with an emphasis on AD and PD, and we discuss iron chelation as a potential therapy. This review also highlights the changes in iron homeostasis caused by NO. An understanding of these mechanisms will help us formulate strategies to reverse or ameliorate iron-related neurodegeneration in diseases such as AD and PD.
Collapse
Affiliation(s)
- Chao Liu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- Neurobiology/Ageing Program, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - Mui Cheng Liang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology/Ageing Program, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology/Ageing Program, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
| |
Collapse
|
6
|
Abstract
As we gain a greater understanding of acne pathogenesis, both new agents as well as new uses for established drugs are being considered for the treatment of acne vulgaris. Multiple clinical trials assessing new formulations or combinations of established acne treatments have been conducted, and novel uses of antimicrobials such as modified diallyl disulfide oxide and nitric oxide are being assessed in clinical trials. There are also a multitude of new therapies currently being studied that target the inflammatory cascade of acne pathogenesis, including sebosuppressive and anti-inflammatory phytochemicals, and small molecule inhibitors targeting sebaceous glands and enzymes, among others. Laser and light therapy is also being modified for the treatment of acne through combination methods with metal nanoshells and vacuum assistance. Probiotics have gained popularity in medicine as greater knowledge of the microbiome and its effects on multiple organ systems is being elucidated. Studies describing the positive effects of certain ammonia-oxidizing bacterial strains in the regulation of the skin's inflammatory response are ongoing. Therapies for acne are constantly evolving and current gold-standard acne therapy may be supplemented with novel treatment modalities in the near future.
Collapse
Affiliation(s)
- Megha K Trivedi
- School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Suzana S Bosanac
- School of Medicine, University of California-Davis, Sacramento, CA, USA
| | - Raja K Sivamani
- Department of Dermatology, University of California, Davis, 3301 C Street, Suite 1400, Sacramento, CA, 95816, USA
- Department of Biological Sciences, California State University, Sacramento, CA, USA
| | - Larissa N Larsen
- Department of Dermatology, University of California, Davis, 3301 C Street, Suite 1400, Sacramento, CA, 95816, USA.
| |
Collapse
|
7
|
Kang DH, Kang OH, Li Z, Mun SH, Seo YS, Kong R, Tian Z, Liu X, Kwon DY. Anti‑inflammatory effects of Ciwujianoside C3, extracted from the leaves of Acanthopanax henryi (Oliv.) Harms, on LPS‑stimulated RAW 264.7 cells. Mol Med Rep 2016; 14:3749-58. [PMID: 27600484 DOI: 10.3892/mmr.2016.5710] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/04/2016] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the unknown mechanisms underlying the anti‑inflammatory activity of Ciwujianoside C3 (CJS C3), extracted from the leaves of Acanthopanax henryi Harms, on lipopolysaccharide (LPS)‑stimulated RAW 264.7 cells. Cells were treated with CJS C3 for 1 h prior to the addition of 200 ng/ml LPS. Cell viability was measured using the MTS assay. Nitric oxide levels were determined by Griess assay. Proinflammatory cytokine production was measured by enzyme‑linked immunosorbent assay. The expression levels of cyclooxygenase (COX)‑2, inducible nitric oxide synthase (iNOS), and mitogen‑activated protein kinases (MAPKs) were investigated by western blotting, reverse transcription (RT)‑polymerase chain reaction (PCR) and RT‑quantitative PCR. Nuclear factor (NF)‑κB/p65 localization, and interaction of the TLR4 receptor with LPS was examined by immunofluorescence assay. The results indicated that CJS C3 exhibited no cytotoxicity at the measured concentrations. Treatment with CJS C3 inhibited NO production, proinflammatory cytokine levels, including interleukin (IL)‑6, tumor necrosis factor (TNF)‑α, and prostaglandin E2 (PGE2), and protein and mRNA expression levels of iNOS and COX‑2. Furthermore, CJS C3 suppressed phosphorylation of extracellular signal‑regulated kinases and c‑jun N‑terminal kinases. It was also able to suppress activation of NF‑κB via inhibition of the TLR4 signaling pathway. These results suggested that CJS C3 exerts inhibitory effects on LPS‑induced PGE2, NO, IL‑6 and TNF‑α production. In addition, iNOS and COX‑2 expression was decreased in murine macrophages. These inhibitory effects may be achieved via suppression of MAPKs and NF‑κB phosphorylation following inhibition of the TLR4 signaling pathway.
Collapse
Affiliation(s)
- Da-Hye Kang
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang‑Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Ok-Hwa Kang
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang‑Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Zhi Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Su-Hyun Mun
- BK21 Plus Team, College of Oriental Medicine, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Yun-Soo Seo
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang‑Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Ryong Kong
- BK21 Plus Team, College of Oriental Medicine, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Zhou Tian
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang‑Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Xiangqian Liu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Dong-Yeul Kwon
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang‑Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| |
Collapse
|
8
|
Vallejos-Vidal E, Reyes-López F, Teles M, MacKenzie S. The response of fish to immunostimulant diets. FISH & SHELLFISH IMMUNOLOGY 2016; 56:34-69. [PMID: 27389620 DOI: 10.1016/j.fsi.2016.06.028] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/25/2016] [Accepted: 06/21/2016] [Indexed: 06/06/2023]
Abstract
In order to maintain fish health and to improve performance immunostimulants have been used as dietary additives to improve weight gain, feed efficiency, and/or disease resistance in cultured fish. In aquaculture, non-specific immunostimulants have been widely used probably due to the limited knowledge of the immune response in fish and the ease of their application. Many studies have been carried out to assess the effect of dietary immunostimulants in fish including algal derivatives, herb and plant extract containing diets using a wide range of downstream analytical techniques. Many immunostimulants are based upon tradition and folklore transferred through generations and specific to certain geographical regions rather than known biological properties. However, there are studies in which it is possible to observe a clear and direct dose-dependent stimulatory effect upon the immune system. Other dietary supplements used contain PAMPs (Pathogen Associated Molecular Patterns) as immunostimulants whose recognition depends upon PRR (pathogen recognition receptor) interactions including the TLRs (Toll-like receptor). Despite the growing interest in the use of immunostimulants across the aquaculture industry the underlying mechanisms of ligand recognition, extract composition and activation of the fish immune response remains fragmented. In this review we focus upon the last 15 years of studies addressing the assessment of: (1) plant, herb and algae extracts; and (2) PAMPs, upon non-specific immune parameters of activation and immunostimulant diet efficacy.
Collapse
Affiliation(s)
- Eva Vallejos-Vidal
- Institut de Biotecnologia i Biomedicina, Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain
| | - Felipe Reyes-López
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain
| | - Mariana Teles
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain
| | - Simon MacKenzie
- Institute of Aquaculture, University of Stirling, FK9 4LA Stirling, UK.
| |
Collapse
|
9
|
Shen P, Fillatreau S. Antibody-independent functions of B cells: a focus on cytokines. Nat Rev Immunol 2015; 15:441-51. [PMID: 26065586 DOI: 10.1038/nri3857] [Citation(s) in RCA: 338] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cytokine production by B cells is important for multiple aspects of immunity. B cell-derived cytokines, including lymphotoxin, are essential for the ontogenesis, homeostasis and activation of secondary lymphoid organs, as well as for the development of tertiary lymphoid tissues at ectopic sites. Other B cell-derived cytokines, such as interleukin-6 (IL-6), interferon-γ and tumour necrosis factor, influence the development of effector and memory CD4(+) T cell responses. Finally, B cells can regulate inflammatory immune responses, primarily through their provision of IL-10 and IL-35. This Review summarizes these various roles of cytokine-producing B cells in immunity and discusses the rational for targeting these cells in the clinic.
Collapse
Affiliation(s)
- Ping Shen
- Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Chariteplatz 1, 10117 Berlin, Germany
| | - Simon Fillatreau
- Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Chariteplatz 1, 10117 Berlin, Germany
| |
Collapse
|
10
|
Pereira J, Porto-Figueira P, Cavaco C, Taunk K, Rapole S, Dhakne R, Nagarajaram H, Câmara JS. Breath analysis as a potential and non-invasive frontier in disease diagnosis: an overview. Metabolites 2015; 5:3-55. [PMID: 25584743 PMCID: PMC4381289 DOI: 10.3390/metabo5010003] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/12/2014] [Indexed: 02/06/2023] Open
Abstract
Currently, a small number of diseases, particularly cardiovascular (CVDs), oncologic (ODs), neurodegenerative (NDDs), chronic respiratory diseases, as well as diabetes, form a severe burden to most of the countries worldwide. Hence, there is an urgent need for development of efficient diagnostic tools, particularly those enabling reliable detection of diseases, at their early stages, preferably using non-invasive approaches. Breath analysis is a non-invasive approach relying only on the characterisation of volatile composition of the exhaled breath (EB) that in turn reflects the volatile composition of the bloodstream and airways and therefore the status and condition of the whole organism metabolism. Advanced sampling procedures (solid-phase and needle traps microextraction) coupled with modern analytical technologies (proton transfer reaction mass spectrometry, selected ion flow tube mass spectrometry, ion mobility spectrometry, e-noses, etc.) allow the characterisation of EB composition to an unprecedented level. However, a key challenge in EB analysis is the proper statistical analysis and interpretation of the large and heterogeneous datasets obtained from EB research. There is no standard statistical framework/protocol yet available in literature that can be used for EB data analysis towards discovery of biomarkers for use in a typical clinical setup. Nevertheless, EB analysis has immense potential towards development of biomarkers for the early disease diagnosis of diseases.
Collapse
Affiliation(s)
- Jorge Pereira
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, Funchal 9000-390, Portugal.
| | - Priscilla Porto-Figueira
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, Funchal 9000-390, Portugal.
| | - Carina Cavaco
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, Funchal 9000-390, Portugal.
| | - Khushman Taunk
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Srikanth Rapole
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Rahul Dhakne
- Laboratory of Computational Biology, Centre for DNA Fingerprinting & Diagnostics, Hyderabad, Andhra Pradesh 500 001, India.
| | - Hampapathalu Nagarajaram
- Laboratory of Computational Biology, Centre for DNA Fingerprinting & Diagnostics, Hyderabad, Andhra Pradesh 500 001, India.
| | - José S Câmara
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, Funchal 9000-390, Portugal.
| |
Collapse
|
11
|
Pham GH, Iglesias BV, Gosselin EJ. Fc receptor-targeting of immunogen as a strategy for enhanced antigen loading, vaccination, and protection using intranasally administered antigen-pulsed dendritic cells. Vaccine 2014; 32:5212-20. [PMID: 25068496 DOI: 10.1016/j.vaccine.2014.07.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 05/14/2014] [Accepted: 07/15/2014] [Indexed: 12/17/2022]
Abstract
Dendritic cells (DCs) play a critical role in the generation of adaptive immunity via the efficient capture, processing, and presentation of antigen (Ag) to naïve T cells. Administration of Ag-pulsed DCs is also an effective strategy for enhancing immunity to tumors and infectious disease organisms. Studies have also demonstrated that targeting Ags to Fcγ receptors (FcγR) on Ag presenting cells can enhance humoral and cellular immunity in vitro and in vivo. Specifically, our studies using a Francisella tularensis (Ft) infectious disease vaccine model have demonstrated that targeting immunogens to FcγR via intranasal (i.n.) administration of monoclonal antibody (mAb)-inactivated Ft (iFt) immune complexes (ICs) enhances protection against Ft challenge. Ft is the causative agent of tularemia, a debilitating disease of humans and other mammals and a category A biothreat agent for which there is no approved vaccine. Therefore, using iFt Ag as a model immunogen, we sought to determine if ex vivo targeting of iFt to FcγR on DCs would enhance the potency of i.n. administered iFt-pulsed DCs. In this study, bone marrow-derived DCs (BMDCs) were pulsed ex vivo with iFt or mAb-iFt ICs. Intranasal administration of mAb-iFt-pulsed BMDCs enhanced humoral and cellular immune responses, as well as protection against Ft live vaccine strain (LVS) challenge. Increased protection correlated with increased iFt loading on the BMDC surface as a consequence of FcγR-targeting. However, the inhibitory FcγRIIB had no impact on this enhancement. In conclusion, targeting Ag ex vivo to FcγR on DCs provides a method for enhanced Ag loading of DCs ex vivo, thereby reducing the amount of Ag required, while also avoiding the inhibitory impact of FcγRIIB. Thus, this represents a simple and less invasive strategy for increasing the potency of ex vivo-pulsed DC vaccines against chronic infectious diseases and cancer.
Collapse
Affiliation(s)
- Giang H Pham
- Center for Immunology and Microbial Disease, 47 New Scotland Avenue, MC-151, Albany Medical College, Albany, NY 12208, United States
| | - Bibiana V Iglesias
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Edmund J Gosselin
- Center for Immunology and Microbial Disease, 47 New Scotland Avenue, MC-151, Albany Medical College, Albany, NY 12208, United States.
| |
Collapse
|
12
|
Bernardino S, Pina A, Felonato M, Costa TA, Frank de Araújo E, Feriotti C, Bazan SB, Keller AC, Leite KRM, Calich VLG. TNF-α and CD8+ T cells mediate the beneficial effects of nitric oxide synthase-2 deficiency in pulmonary paracoccidioidomycosis. PLoS Negl Trop Dis 2013; 7:e2325. [PMID: 23936574 PMCID: PMC3731220 DOI: 10.1371/journal.pntd.0002325] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 06/09/2013] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Nitric oxide (NO), a key antimicrobial molecule, was previously shown to exert a dual role in paracoccidioidomycosis, an endemic fungal infection in Latin America. In the intravenous and peritoneal models of infection, NO production was associated with efficient fungal clearance but also with non-organized granulomatous lesions. Because paracoccidioidomycosis is a pulmonary infection, we aimed to characterize the role of NO in a pulmonary model of infection. METHODOLOGY/PRINCIPAL FINDINGS C57Bl/6 wild type (WT) and iNOS(-/-) mice were i.t. infected with 1×10(6) Paracoccidioides brasiliensis yeasts and studied at several post-infection periods. Unexpectedly, at week 2 of infection, iNOS(-/-) mice showed decreased pulmonary fungal burdens associated with an M2-like macrophage profile, which expressed high levels of TGF-β impaired ability of ingesting fungal cells. This early decreased fungal loads were concomitant with increased DTH reactions, enhanced TNF-α synthesis and intense migration of activated macrophages, CD4(+) and CD8(+) T cells into the lungs. By week 10, iNOS(-/-) mice showed increased fungal burdens circumscribed, however, by compact granulomas containing elevated numbers of activated CD4(+) T cells. Importantly, the enhanced immunological reactivity of iNOS(-/-) mice resulted in decreased mortality rates. In both mouse strains, depletion of TNF-α led to non-organized lesions and excessive influx of inflammatory cells into the lungs, but only the iNOS(-/-) mice showed increased mortality rates. In addition, depletion of CD8(+) cells abolished the increased migration of inflammatory cells and decreased the number of TNF-α and IFN-γ CD4(+) and CD8(+) T cells into the lungs of iNOS(-/-) mice. CONCLUSIONS/SIGNIFICANCE Our study demonstrated that NO plays a deleterious role in pulmonary paracoccidioidomycosis due to its suppressive action on TNF-α production, T cell immunity and organization of lesions resulting in precocious mortality of mice. It was also revealed that uncontrolled fungal growth can be overcome by an efficient immune response.
Collapse
Affiliation(s)
- Simone Bernardino
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| | - Adriana Pina
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| | - Maíra Felonato
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| | - Tânia A. Costa
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| | - Eliseu Frank de Araújo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| | - Cláudia Feriotti
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| | - Silvia Boschi Bazan
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| | - Alexandre C. Keller
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, Brasil
| | - Katia R. M. Leite
- Departamento de Patologia, Hospital Sírio Libanês de São Paulo, São Paulo, Brasil
| | - Vera L. G. Calich
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| |
Collapse
|
13
|
Abstract
Francisella tularensis is a gram-negative bacterium that causes the zoonotic disease tularemia. Francisella is highly infectious via the respiratory route (~10 CFUs) and pulmonary infections due to type A strains of F. tularensis are highly lethal in untreated patients (>30%). In addition, no vaccines are licensed to prevent tularemia in humans. Due to the high infectivity and mortality of pulmonary tularemia, F. tularensis has been weaponized, including via the introduction of antibiotic resistance, by several countries. Because of the lack of efficacious vaccines, and concerns about F. tularensis acquiring resistance to antibiotics via natural or illicit means, augmentation of host immunity, and humoral immunotherapy have been investigated as countermeasures against tularemia. This manuscript will review advances made and challenges in the field of immunotherapy against tularemia.
Collapse
Affiliation(s)
- Jerod A Skyberg
- Department of Veterinary Pathobiology and Laboratory for Infectious Disease Research; University of Missouri; Columbia, MO USA
| |
Collapse
|
14
|
Szczytkowski JL, Lebonville C, Hutson L, Fuchs RA, Lysle DT. Heroin-induced conditioned immunomodulation requires expression of IL-1β in the dorsal hippocampus. Brain Behav Immun 2013; 30:95-102. [PMID: 23357470 PMCID: PMC3641184 DOI: 10.1016/j.bbi.2013.01.076] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/14/2013] [Accepted: 01/14/2013] [Indexed: 12/31/2022] Open
Abstract
Opioid-associated environmental stimuli elicit robust immune-altering effects via stimulation of a neural circuitry that includes the basolateral amygdala and nucleus accumbens. These brain regions are known to have both direct and indirect connections with the hippocampus. Thus, the present study evaluated whether the dorsal hippocampus (DH), and more specifically interleukin-1 beta (IL-1β) within the DH, is necessary for the expression of heroin-induced conditioned immunomodulation. Rats received five Pavlovian pairings of systemic heroin administration (1.0mg/kg, SC) with placement into a distinct environment (conditioned stimulus, CS). Six days after conditioning, a GABAA/B agonist cocktail or IL-1β small interfering RNA (siRNA) was microinfused into the DH to inhibit neuronal activity or IL-1β gene expression prior to CS or home cage exposure. Control animals received saline or negative control siRNA microinfusions. Furthermore, all rats received systemic administration of lipopolysaccharide (LPS) to stimulate proinflammatory nitric oxide production. CS exposure suppressed LPS-induced nitric oxide production relative to home cage exposure. Inactivation of, or IL-1β silencing in, the DH disrupted the CS-induced suppression of nitric oxide production relative to vehicle or negative control siRNA treatment. These results are the first to show a role for DH IL-1β expression in heroin-conditioned suppression of a proinflammatory immune response.
Collapse
Affiliation(s)
- Jennifer L. Szczytkowski
- University of North Carolina at Chapel Hill, Department of Psychology, CB#3270, Chapel Hill, NC 27599-3270 USA
- Messiah College, Department of Psychology, One College Avenue Suite 3052, Mechanicsburg, PA 17055 USA
| | - Christina Lebonville
- University of North Carolina at Chapel Hill, Department of Psychology, CB#3270, Chapel Hill, NC 27599-3270 USA
| | - Lee Hutson
- University of North Carolina at Chapel Hill, Department of Psychology, CB#3270, Chapel Hill, NC 27599-3270 USA
| | - Rita A. Fuchs
- University of North Carolina at Chapel Hill, Department of Psychology, CB#3270, Chapel Hill, NC 27599-3270 USA
| | - Donald T. Lysle
- University of North Carolina at Chapel Hill, Department of Psychology, CB#3270, Chapel Hill, NC 27599-3270 USA
| |
Collapse
|
15
|
Ashtekar AR, Katz J, Xu Q, Michalek SM. A mucosal subunit vaccine protects against lethal respiratory infection with Francisella tularensis LVS. PLoS One 2012; 7:e50460. [PMID: 23209745 PMCID: PMC3508931 DOI: 10.1371/journal.pone.0050460] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 10/22/2012] [Indexed: 01/18/2023] Open
Abstract
Francisella tularensis (FT) is a highly virulent pathogen for humans and other mammals. Severe morbidity and mortality is associated with respiratory FT infection and there are concerns about intentional dissemination of this organism. Therefore, FT has been designated a category A biothreat agent and there is a growing interest in the development of a protective vaccine. In the present study, we determine the protective potential of a subunit vaccine comprised of the FT heat shock protein DnaK and surface lipoprotein Tul4 against respiratory infection with the live vaccine strain (LVS) of FT in mice. First, we establish an optimal intranasal immunization regimen in C57BL/6 mice using recombinant DnaK or Tul4 together with the adjuvant GPI-0100. The individual immunization regimens induced robust salivary IgA, and vaginal and bronchoalveolar IgA and IgG antigen-specific antibodies. Serum IgG1 and IgG2c antibody responses were also induced, indicative of a mixed type 2 and type 1 response, respectively. Next, we show that immunization with DnaK and Tul4 induces mucosal and systemic antibody responses that are comparable to that seen following immunization with each antigen alone. This immunization regimen also induced IFN-γ, IL-10 and IL-17A production by splenic CD4(+) T cells in an antigen-specific manner. Importantly, over 80% of the mice immunized with DnaK and Tul4, but not with each antigen alone, were protected against a lethal respiratory challenge with FT LVS. Protection correlated with reduced bacterial burden in the lung, liver and spleen of mice. This study demonstrates the potential of DnaK and Tul4 as protective antigens and lends support to the notion of combining distinct, immunodominant antigens into an effective multivalent tularemia vaccine.
Collapse
Affiliation(s)
- Amit R. Ashtekar
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jannet Katz
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Qingan Xu
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Suzanne M. Michalek
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
16
|
Cole LE, Mann BJ, Shirey KA, Richard K, Yang Y, Gearhart PJ, Chesko KL, Viscardi RM, Vogel SN. Role of TLR signaling in Francisella tularensis-LPS-induced, antibody-mediated protection against Francisella tularensis challenge. J Leukoc Biol 2011; 90:787-97. [PMID: 21750122 DOI: 10.1189/jlb.0111014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Immunization with Ft-LPS provokes an antigen-specific, B-1a cell-derived antibody response that protects WT mice against an otherwise lethal challenge with Ft LVS. However, this same regimen offers limited protection to TLR2(-/-) mice, despite production of WT levels of anti-Ft-LPS antibodies. As Ft-LPS exhibits no TLR2 agonist activity, and macrophage-induced cytokine production in response to Ft LVS is overwhelmingly TLR2-dependent, we hypothesized that treatment of TLR2(-/-) mice with an alternative, MyD88-dependent TLR agonist would compensate for reduced recognition of Ft LVS in TLR2(-/-) mice and thereby, restore Ft-LPS-mediated protection. Administration of the nontoxic TLR4 agonist, synthetic Escherichia coli MPL, at the time of Ft-LPS immunization or Ft LVS challenge, fully protected TLR2(-/-) mice, whereas treatment of WT or TLR2(-/-) mice with MPL alone conferred partial protection. The TLR5 agonist, flagellin, also synergized with Ft-LPS to protect TLR2(-/-) mice from lethal Ft LVS challenge. In contrast to Ft LVS, Ft-LPS pretreatment failed to protect mice against i.n. challenge with Ft Schu S4, whereas MPL, administered in the absence or presence of Ft-LPS, conferred significant, albeit partial, protection. MPL treatment of macrophages increased the uptake of Ft LVS and decreased intracellular bacterial survival while shifting the macrophage-differentiation phenotype from "alternatively activated" to "classically activated". Collectively, our data suggest that optimal, Ft-LPS-mediated protection against Ft LVS infection requires two discrete events, i.e., production of Ft-LPS-specific antibody, as well as TLR-mediated macrophage activation, to fully control Francisella infection.
Collapse
Affiliation(s)
- Leah E Cole
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Fillatreau S. Novel regulatory functions for Toll-like receptor-activated B cells during intracellular bacterial infection. Immunol Rev 2011; 240:52-71. [PMID: 21349086 DOI: 10.1111/j.1600-065x.2010.00991.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Infections by intracellular bacterial pathogens remain a major cause of human diseases worldwide. Despite intensive efforts, the development of effective vaccines or immunotherapies against these diseases has largely remained unsuccessful, asking for the exploration of new aspects of the host response to these pathogens. Genetic studies have demonstrated beyond doubt that cell-mediated mechanisms of host defense involving innate immunity and T cells are of crucial importance for the control of these diseases. By contrast, the role of B cells during intracellular bacterial infection has so far received little attention besides their role as antibody-producing cells. However, the general knowledge of B-cell immunology and in particular of their antibody-independent functions has greatly increased during the last years. Recently, it was found in a model of Salmonella typhimurium infection that Toll-like receptor triggering on B cells resulted through interleukin-10 secretion in a marked suppression of innate defense mechanisms ultimately leading to uncontrolled growth of the bacteria and earlier death from the disease during both primary and secondary infections. This article reviews the protective and deleterious roles of B cells during intracellular bacterial infections and discusses how manipulating their antibody-independent functions may be a powerful means to therapeutically improve host resistance against these diseases.
Collapse
Affiliation(s)
- Simon Fillatreau
- Deutsches Rheuma-ForschungsZentrum, Leibniz Institute, Berlin, Germany.
| |
Collapse
|
18
|
Lipscomb MF, Hutt J, Lovchik J, Wu T, Lyons CR. The pathogenesis of acute pulmonary viral and bacterial infections: investigations in animal models. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2010; 5:223-52. [PMID: 19824827 DOI: 10.1146/annurev-pathol-121808-102153] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Acute viral and bacterial infections in the lower respiratory tract are major causes of morbidity and mortality worldwide. The proper study of pulmonary infections requires interdisciplinary collaboration among physicians and biomedical scientists to develop rational hypotheses based on clinical studies and to test these hypotheses in relevant animal models. Animal models for common lung infections are essential to understand pathogenic mechanisms and to clarify general mechanisms for host protection in pulmonary infections, as well as to develop vaccines and therapeutics. Animal models for uncommon pulmonary infections, such as those that can be caused by category A biothreat agents, are also very important because the infrequency of these infections in humans limits in-depth clinical studies. This review summarizes our understanding of innate and adaptive immune mechanisms in the lower respiratory tract and discusses how animal models for selected pulmonary pathogens can contribute to our understanding of the pathogenesis of lung infections and to the search for new vaccines and therapies.
Collapse
Affiliation(s)
- Mary F Lipscomb
- Departments of Pathology and University of New Mexico School of Medicine, Albuquerque, New Mexico 87131.
| | | | | | | | | |
Collapse
|
19
|
Identification of Francisella tularensis live vaccine strain CuZn superoxide dismutase as critical for resistance to extracellularly generated reactive oxygen species. J Bacteriol 2009; 191:6447-56. [PMID: 19684141 DOI: 10.1128/jb.00534-09] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Francisella tularensis is an intracellular pathogen whose survival is in part dependent on its ability to resist the microbicidal activity of host-generated reactive oxygen species (ROS) and reactive nitrogen species (RNS). In numerous bacterial pathogens, CuZn-containing superoxide dismutases (SodC) are important virulence factors, localizing to the periplasm to offer protection from host-derived superoxide radicals (O(2)(-)). In the present study, mutants of F. tularensis live vaccine strain (LVS) deficient in superoxide dismutases (SODs) were used to examine their role in defense against ROS/RNS-mediated microbicidal activity of infected macrophages. An in-frame deletion F. tularensis mutant of sodC (DeltasodC) and a F. tularensis DeltasodC mutant with attenuated Fe-superoxide dismutase (sodB) gene expression (sodB DeltasodC) were constructed and evaluated for susceptibility to ROS and RNS in gamma interferon (IFN-gamma)-activated macrophages and a mouse model of respiratory tularemia. The F. tularensis DeltasodC and sodB DeltasodC mutants showed attenuated intramacrophage survival in IFN-gamma-activated macrophages compared to the wild-type F. tularensis LVS. Transcomplementing the sodC gene in the DeltasodC mutant or inhibiting the IFN-gamma-dependent production of O(2)(-) or nitric oxide (NO) enhanced intramacrophage survival of the sod mutants. The DeltasodC and sodB DeltasodC mutants were also significantly attenuated for virulence in intranasally challenged C57BL/6 mice compared to the wild-type F. tularensis LVS. As observed for macrophages, the virulence of the DeltasodC mutant was restored in ifn-gamma(-/-), inos(-/-), and phox(-/-) mice, indicating that SodC is required for resisting host-generated ROS. To conclude, this study demonstrates that SodB and SodC act to confer protection against host-derived oxidants and contribute to intramacrophage survival and virulence of F. tularensis in mice.
Collapse
|
20
|
Contribution of citrulline ureidase to Francisella tularensis strain Schu S4 pathogenesis. J Bacteriol 2009; 191:4798-806. [PMID: 19502406 DOI: 10.1128/jb.00212-09] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The citrulline ureidase (CTU) activity has been shown to be associated with highly virulent Francisella tularensis strains, including Schu S4, while it is absent in avirulent or less virulent strains. A definitive role of the ctu gene in virulence and pathogenesis of F. tularensis Schu S4 has not been assessed; thus, an understanding of the significance of this phenotype is long overdue. CTU is a carbon-nitrogen hydrolase encoded by the citrulline ureidase (ctu) gene (FTT0435) on the F. tularensis Schu S4 genome. In the present study, we evaluated the contribution of the ctu gene in the virulence of category A agent F. tularensis Schu S4 by generating a nonpolar deletion mutant, the Deltactu mutant. The deletion of the ctu gene resulted in loss of CTU activity, which was restored by transcomplementing the ctu gene. The Deltactu mutant did not exhibit any growth defect under acellular growth conditions; however, it was impaired for intramacrophage growth in resting as well as gamma interferon-stimulated macrophages. The Deltactu mutant was further tested for its virulence attributes in a mouse model of respiratory tularemia. Mice infected intranasally with the Deltactu mutant showed significantly reduced bacterial burden in the lungs, liver, and spleen compared to wild-type (WT) Schu S4-infected mice. The reduced bacterial burden in mice infected with the Deltactu mutant was also associated with significantly lower histopathological scores in the lungs. Mice infected with the Deltactu mutant succumbed to infection, but they survived longer and showed significantly extended median time to death compared to that shown by WT Schu S4-infected mice. To conclude, this study demonstrates that ctu contributes to intracellular survival, in vivo growth, and pathogenesis. However, ctu is not an absolute requirement for the virulence of F. tularensis Schu S4 in mice.
Collapse
|
21
|
Mucosal immunotherapy for protection from pneumonic infection with Francisella tularensis. Vaccine 2009; 27:4424-33. [PMID: 19490961 DOI: 10.1016/j.vaccine.2009.05.041] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 05/08/2009] [Accepted: 05/12/2009] [Indexed: 12/11/2022]
Abstract
Previous studies have demonstrated that systemically administered immunotherapy can protect mice from systemic challenge with the bacterial pathogen Francisella tularensis. However, for protection from inhalational challenge with this bacterium, we wondered if mucosally administered immunotherapy might be more effective. Therefore, we administered cationic liposome-DNA complexes (CLDC), which are potent activators of innate immunity, intranasally (i.n.) and assessed the effectiveness of protection from lethal inhalational challenge with F. tularensis. We found that pretreatment by i.n. administration of CLDC 24h prior to bacterial challenge elicited nearly complete protection of BALB/c mice from lethal challenge with F. tularensis LVS strain. We also observed that mucosal CLDC immunotherapy provided a statistically significant increase in survival time in mice challenged with the highly virulent F. tularensis Schu4 strain. Protection was associated with a significant reduction in bacterial burden in the lungs, liver, and spleen. Mucosal administration of CLDC elicited significantly increased expression of IL-12, IFN-gamma, TNF-alpha, IFN-beta and IFN-alpha genes in the lung as detected by real-time quantitative PCR. In vitro treatment of F. tularensis infected macrophages with CLDC-elicited cytokines also significantly suppressed intracellular replication of F. tularensis in infected macrophages. In vivo, depletion of NK cells prior to administration of CLDC completely abolished the protective effects of CLDC immunotherapy. CLDC-elicited protection was also dependent on induction of IFN-gamma production in vivo. We conclude therefore that activation of local pulmonary innate immune responses is capable of eliciting significant protection from inhalational exposure to a virulent bacterial pathogen.
Collapse
|
22
|
Bitsaktsis C, Rawool DB, Li Y, Kurkure NV, Iglesias B, Gosselin EJ. Differential requirements for protection against mucosal challenge with Francisella tularensis in the presence versus absence of cholera toxin B and inactivated F. tularensis. THE JOURNAL OF IMMUNOLOGY 2009; 182:4899-909. [PMID: 19342669 DOI: 10.4049/jimmunol.0803242] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Francisella tularensis is a category A biothreat agent for which there is no approved vaccine and the correlates of protection are not well understood. In particular, the relationship between the humoral and cellular immune response to F. tularensis and the relative importance of each in protection is controversial. Yet, understanding this relationship will be crucial to the development of an effective vaccine against this organism. We demonstrate, for the first time, a differential requirement for humoral vs cellular immunity in vaccine-induced protection against F. tularensis infection, and that the requirement for Ab observed in some protection studies, may be overcome through the induction of enhanced cellular immunity. Specifically, following intranasal/mucosal immunization of mice with inactivated F. tularensis organisms plus the cholera toxin B subunit, we observe increased production of IgG2a/2c vs IgG1 Ab, as well as IFN-gamma, indicating induction of a Th1 response. In addition, the requirement for F. tularensis-specific IgA Ab production, observed in studies following immunization with inactivated F. tularensis alone, is eliminated. Thus, these data indicate that enhanced Th1 responses can supersede the requirement for anti-F. tularensis-specific IgA. This observation also has important ramifications for vaccine development against this organism.
Collapse
Affiliation(s)
- Constantine Bitsaktsis
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | | | | | | | | | | |
Collapse
|
23
|
Kirimanjeswara GS, Golden JM, Bakshi CS, Metzger DW. Prophylactic and Therapeutic Use of Antibodies for Protection against Respiratory Infection withFrancisella tularensis. THE JOURNAL OF IMMUNOLOGY 2007; 179:532-9. [PMID: 17579074 DOI: 10.4049/jimmunol.179.1.532] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The role of Abs in protection against respiratory infection with the intracellular bacterium Francisella tularensis is not clear. To investigate the ability of Abs to clear bacteria from the lungs and prevent systemic spread, immune serum was passively administered i.p. to naive mice before intranasal F. tularensis live vaccine strain infection. It was found that immune serum treatment provided 100% protection against lethal challenge while normal serum or Ig-depleted immune serum provided no protection. Protective efficacy was correlated with increased clearance of bacteria from the lung and required expression of FcgammaR on phagocytes, including macrophages and neutrophils. However, complement was not required for protection. In vitro experiments demonstrated that macrophages were more readily infected by Ab-opsonized bacteria but became highly efficient in killing upon activation by IFN-gamma. Consistent with this finding, in vivo Ab-mediated protection was found to be dependent upon IFN-gamma. SCID mice were not protected by passive Ab transfer, suggesting that T cells but not NK cells serve as the primary source for IFN-gamma. These data suggest that a critical interaction of humoral and cellular immune responses is necessary to provide sterilizing immunity against F. tularensis. Of considerable interest was the finding that serum Abs were capable of conferring protection against lethal respiratory tularemia when given 24-48 h postexposure. Thus, this study provides the first evidence for the therapeutic use of Abs in Francisella-infected individuals.
Collapse
Affiliation(s)
- Girish S Kirimanjeswara
- Center for Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Avenue Albany, NY 12208, USA
| | | | | | | |
Collapse
|
24
|
Basagoudanavar SH, Goswami PP, Tiwari V. Cellular immune responses to 35 kDa recombinant antigen of Mycobacterium avium paratuberculosis. Vet Res Commun 2007; 30:357-67. [PMID: 16502104 DOI: 10.1007/s11259-006-3253-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2004] [Indexed: 10/25/2022]
Abstract
Mycobacterium avium paratuberculosis is the causative agent of Johne disease, a chronic ulcerative intestinal condition in ruminant animals. Owing to the predominance of cellular response in subclinical forms of the infection, identification of M. a. paratuberculosis antigens eliciting host cell-mediated immune (CMI) reaction is crucial for early control of the disease. A 35 kDa protein of M. a. paratuberculosis was studied for its ability to elicit CMI responses using a mouse model. Lymphoproliferation and IFN-gamma response were used to measure the CMI response. Recombinant 35 kDa protein (P35) stimulated proliferation of mouse mononuclear splenocytes sensitized with M. a. paratuberculosis. The P35 elicited increased nitrite production from mononuclear splenocytes from M. a. paratuberculosis-sensitized mice. In addition, RT-PCR-based semiquantitative IFN-gamma measurement showed that stimulation with P35 is associated with significant expression of IFN-gamma mRNA in M. a. paratuberculosis-sensitized mouse splenocytes. The results indicate that the 35 kDa protein of M. a. paratuberculosis is associated with CMI response in the host.
Collapse
Affiliation(s)
- S H Basagoudanavar
- National Biotechnology Centre, Indian Veterinary Research Institute (IVRI), Izatnagar, 243 122, India
| | | | | |
Collapse
|
25
|
McLendon MK, Apicella MA, Allen LAH. Francisella tularensis: taxonomy, genetics, and Immunopathogenesis of a potential agent of biowarfare. Annu Rev Microbiol 2006; 60:167-85. [PMID: 16704343 PMCID: PMC1945232 DOI: 10.1146/annurev.micro.60.080805.142126] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Tularemia is a zoonosis of humans caused by infection with the facultative intracellular bacterium Francisella tularensis. Interest in F. tularensis has increased markedly in the past few years because of its potential use as an agent of bioterrorism. Five subspecies of this organism are found in the Northern hemisphere, but only F. tularensis subsp. tularensis and subsp. holarctica cause disease in humans. This review summarizes what is known about the pathogenesis of tularemia with a focus on bacterial surface components such as lipopolysaccharide and capsule as well as information obtained from the F. tularensis subsp. tularensis SCHU S4 genome. In particular, the mechanisms of action of recently identified virulence factors are discussed in the context of bacterial replication in macrophages and manipulation of the host inflammatory response. Throughout this report, shared and unique features of F. tularensis subsp. tularensis, subsp. holarctica, and subsp. novicida are discussed.
Collapse
Affiliation(s)
- Molly K McLendon
- Inflammation Program, Department of Microbiology, University of Iowa and the VA Medical Center, Iowa City, Iowa 52242, USA.
| | | | | |
Collapse
|
26
|
Francisella tularensis LVS grown in macrophages has reduced ability to stimulate the secretion of inflammatory cytokines by macrophages in vitro. Microb Pathog 2006; 41:218-25. [PMID: 16996713 DOI: 10.1016/j.micpath.2006.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Revised: 07/25/2006] [Accepted: 07/27/2006] [Indexed: 02/07/2023]
Abstract
The virulence of Francisella tularensis LVS is determined in part by its ability to invade and replicate within macrophages and stimulate the production of inflammatory cytokines. The present study determined the effects of growing F. tularensis in macrophages on its ability to stimulate cytokine secretion by macrophages. F. tularensis grown in Mueller-Hinton broth (FtB) stimulated the secretion of large amounts of TNF-alpha, IL-12p40, IL-6 and MCP-1/CCL2 when incubated with macrophages overnight. In contrast, F. tularensis released from infected macrophages (FtMac) stimulated very little secretion of these cytokines by primary cultures of murine peritoneal macrophages, human monocytes or macrophage cell lines. Stimulation of nitric oxide production by FtMac was also less than that elicited by FtB. FtMac killed with gentamicin or paraformaldehyde also stimulated low levels of cytokine secretion. FtMac recovered the ability to stimulate cytokine secretion after overnight culture in broth. Infection of macrophages with FtMac inhibited the cytokine response to subsequent stimulation with LPS from Escherichia coli but did not affect Fcgamma receptor-mediated phagocytosis. FtMac were ingested by macrophages at about half the rate of FtB, however, this did not account for the lower cytokine secretion. FtMac and FtB replicated at similar rates within macrophages. Finally, Mice infected with FtMac had a higher mortality rate than those infected with FtB. These results reveal that growth in macrophages causes a reversible phenotypic change in F. tularensis that is associated with decreased stimulation of cytokine secretion, inhibition of LPS-stimulated secretion of inflammatory cytokines by macrophages and increased lethality in mice.
Collapse
|
27
|
Shirey KA, Jung JY, Maeder GS, Carlin JM. Upregulation of IFN-gamma receptor expression by proinflammatory cytokines influences IDO activation in epithelial cells. J Interferon Cytokine Res 2006; 26:53-62. [PMID: 16426148 PMCID: PMC1550344 DOI: 10.1089/jir.2006.26.53] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interferon-gamma (IFN-gamma) induces the enzyme indoleamine dioxygenase (IDO) in a variety of human cell types. Furthermore, tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 (IL-1) synergistically increase IFN-induced IDO activity. Inasmuch as cytokines can upregulate cytokine receptor expression, one mechanism of cytokine synergy may be at the level of receptor expression. To test the hypothesis that this mechanism of IDO regulation is active in epithelial cells, HeLa cells were treated with IFN-gamma, TNF-alpha, or IL-1beta to determine optimal cytokine concentrations and time for maximal cytokine receptor expression. Flow cytometric analysis with antibodies to receptors for IFN-gamma, TNF-alpha, or IL-1beta indicated that each cytokine upregulated expression of the other cytokine receptors by 4 h, with maximal expression observed between 16 and 20 h after cytokine treatment. Furthermore, increases in IFN-gamma receptors (IFNGR) induced by IL-1beta were found to be dependent on NF-kappaB transactivation. To determine if increases in IFNGR expression alone contributes to synergistic IDO induction, cells were stimulated with IL-1beta to upregulate receptor expression, and the NF-kappaB concentration was allowed to return to basal levels. When treated with IFN-gamma, enhanced Stat1 signaling and IDO induction were still observed, indicating that increased cytokine receptor expression contributes to synergistic increases in IDO activity.
Collapse
Affiliation(s)
- Kari Ann Shirey
- Department of Microbiology, Miami University, Oxford, OH 45056, USA
| | | | | | | |
Collapse
|
28
|
Fukuhara M, Fukazawa M, Tamura A, Nakamura T, Urakami H. Survival of two Orientia tsutsugamushi bacterial strains that infect mouse macrophages with varying degrees of virulence. Microb Pathog 2005; 39:177-87. [PMID: 16165341 DOI: 10.1016/j.micpath.2005.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Revised: 08/02/2005] [Accepted: 08/05/2005] [Indexed: 11/23/2022]
Abstract
Orientia tsutsugamushi, an intracellular parasitic bacterium, comprises numerous strains of differing virulence. When BALB/c mice were infected intraperitoneally with this pathogen, a virulent strain known as Karp was found to multiply in the intraperitoneal macrophages and kill the mouse. In contrast, an avirulent strain, Kuroki, was shown to invade macrophages but be eliminated from the cells, allowing mouse survival. O. tsutsugamushi invades its host cell cytoplasm through phagocytosis and disruption of phagosomal membranes but some bacteria are then killed by phago-lysosomes within 1h of infection. Microscopic observations could not differentiate the Karp and Kuroki strains during entry and subsequent cell killing by phago-lysosomes. However, the Kuroki cells failed to divide and were markedly deformed following cytoplasmic invasion at several days post-infection. These findings suggest that macrophages have a mechanism to eliminate O. tsutsugamushi in the cytoplasm, if the invading bacteria escape phagosomal clearance, and that it is this mechanism that Kuroki does not survive. Additionally, significant levels of nitric oxide (NO) are produced in macrophages by Kuroki, but not by Karp. An NO synthase inhibitor, however, does not increase the growth of Kuroki, suggesting that NO is induced in a strain-dependent manner but does not effect proliferation.
Collapse
Affiliation(s)
- Masahiro Fukuhara
- Laboratory of Microbiology, Faculty of Pharmacy, Niigata University of Pharmacy and Applied Life Sciences (NUPALS), 265-1 Higashijima, Niigata 956-8603, Japan
| | | | | | | | | |
Collapse
|
29
|
Leitão RFC, Ribeiro RA, Chaves HV, Rocha FAC, Lima V, Brito GAC. Nitric oxide synthase inhibition prevents alveolar bone resorption in experimental periodontitis in rats. J Periodontol 2005; 76:956-63. [PMID: 15948691 DOI: 10.1902/jop.2005.76.6.956] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Periodontitis is the most frequent cause of tooth loss in adults. Nitric oxide (NO) has been linked to bone resorption mechanisms during inflammation processes. The aim of this study was to investigate the effect of NOS (NO synthase) inhibitors in the alveolar bone loss in an experimental periodontitis disease (EPD) model. METHODS Wistar rats were subjected to a ligature placement around the second upper left molars and were sacrificed at 11 days. Alveolar bone loss was evaluated by the sum of distances between the cusp tips and the alveolar bone along the axis of each molar root, subtracting from the contralateral side. Histopathological analysis was based on cell influx, alveolar bone, and cementum integrity. Leukogram was performed at 6 hours and 1, 7, and 11 days after the EPD induction. Groups were treated with the NOS inhibitors, aminoguanidine (AG) (2.5 to 10 mg/kg/d), or L-arginine methyl ester (L-NAME, 5 to 20 mg/kg/d) intraperitoneally (i.p.), 1 hour before the EPD induction and daily for 11 days. Controls received only saline (EPD group). As controls for L-NAME specificity, groups were co-treated with either L-arginine (150 to 600 mg/kg/d) or D-arginine (600 mg/kg/d) and L-NAME (20 mg/kg/d). Different groups were used for morphometric and histopathological analysis. RESULTS Both L-NAME and AG significantly and dose-dependently inhibited the alveolar bone loss as compared to EPD group. L-NAME (20 mg/kg/d) reduced the alveolar bone loss by 50%, whereas AG (5 mg/kg/d) reduced it by 47% compared to EPD. This result was coupled to a significant reduction of cell influx to the periodontium, as well as to the preservation of alveolar bone and cementum, seen at histopathology, for both compounds. The co-administration of L-arginine, but not of D-arginine reversed L-NAME effects. CONCLUSION These data provide evidence that NOS inhibitors prevent inflammatory bone resorption in experimental periodontitis.
Collapse
Affiliation(s)
- R F C Leitão
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Ceará, Brazil
| | | | | | | | | | | |
Collapse
|
30
|
Mollace V, Muscoli C, Masini E, Cuzzocrea S, Salvemini D. Modulation of prostaglandin biosynthesis by nitric oxide and nitric oxide donors. Pharmacol Rev 2005; 57:217-52. [PMID: 15914468 DOI: 10.1124/pr.57.2.1] [Citation(s) in RCA: 250] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The biosynthesis and release of nitric oxide (NO) and prostaglandins (PGs) share a number of similarities. Two major forms of nitric-oxide synthase (NOS) and cyclooxygenase (COX) enzymes have been identified to date. Under normal circumstances, the constitutive isoforms of these enzymes (constitutive NOS and COX-1) are found in virtually all organs. Their presence accounts for the regulation of several important physiological effects (e.g. antiplatelet activity, vasodilation, and cytoprotection). On the other hand, in inflammatory setting, the inducible isoforms of these enzymes (inducible NOS and COX-2) are detected in a variety of cells, resulting in the production of large amounts of proinflammatory and cytotoxic NO and PGs. The release of NO and PGs by the inducible isoforms of NOS and COX has been associated with the pathological roles of these mediators in disease states as evidenced by the use of selective inhibitors. An important link between the NOS and COX pathways was made in 1993 by Salvemini and coworkers when they demonstrated that the enhanced release of PGs, which follows inflammatory mechanisms, was nearly entirely driven by NO. Such studies raised the possibility that COX enzymes represent important endogenous "receptor" targets for modulating the multifaceted roles of NO. Since then, numerous papers have been published extending the observation across various cellular systems and animal models of disease. Furthermore, other studies have highlighted the importance of such interaction in physiology as well as in the mechanism of action of drugs such as organic nitrates. More importantly, mechanistic studies of how NO switches on/off the PG/COX pathway have been undertaken and additional pathways through which NO modulates prostaglandin production unraveled. On the other hand, NO donors conjugated with COX inhibitors have recently found new interest in the understanding of NO/COX reciprocal interaction and potential clinical use. The purpose of this article is to cover the advances which have occurred over the years, and in particular, to summarize experimental data that outline how the discovery that NO modulates prostaglandin production has impacted and extended our understanding of these two systems in physiopathological events.
Collapse
Affiliation(s)
- Vincenzo Mollace
- Faculty of Pharmacy, University of Catanzaro Magna Graecia, Roccelletta di Borgia, Catanazaro, Italy
| | | | | | | | | |
Collapse
|
31
|
Santic M, Molmeret M, Abu Kwaik Y. Modulation of biogenesis of the Francisella tularensis subsp. novicida-containing phagosome in quiescent human macrophages and its maturation into a phagolysosome upon activation by IFN-gamma. Cell Microbiol 2005; 7:957-67. [PMID: 15953028 DOI: 10.1111/j.1462-5822.2005.00529.x] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Francisella tularensis is a highly virulent facultative intracellular pathogen that has been categorized as a class A bioterrorism agent, and is classified into four subsp, tularensis, holarctica, mediasiatica and novicida. Although the ability of F. tularensis subsp. novicida to cause tularemia in mice is similar to the virulent subsp. tularensis and holarctica, it is attenuated in humans. It is not known whether attenuation of F. tularensis subsp. novicida in humans is resulting from a different route of trafficking within human macrophages, compared with the tularensis or holarctica subsp. Here we show that in quiescent human monocytes-derived macrophages (hMDMs), the F. tularensis subsp. novicida containing phagosome (FCP) matures into a late endosome-like stage that acquires the late endosomal marker LAMP-2 but does not fuse to lysosomes. This modulation of phagosome biogenesis by F. tularensis is followed by disruption of the phagosome at 4-12 h and subsequent bacterial escape into cytoplasm where the organism replicates. In IFN-gamma-activated hMDMs, intracellular replication of F. tularensis is completely inhibited, and is associated with failure of the organism to escape from the phagosome into the cytoplasm for up to 24 h after infection. In IFN-gamma-activated hMDMs, the FCPs acquire the lysosomal enzymes Cathepsin D, which is excluded in quiescent hMDMs. When the lysosomes of IFN-gamma-activated hMDMs are preload with Texas Red Ovalbumin or BSA-gold, the FCPs acquire both lysosomal tracers. In contrast, both lysosomal tracers are excluded from the FCPs within quiescent hMDMs. We conclude that although F. tularensis subsp. novicida is attenuated in humans, it modulates biogenesis of its phagosome into a late endosome-like compartment followed by bacterial escape into the cytoplasm within quiescent hMDMs, similar to the virulent subsp. tularensis. In IFN-gamma-activated hMDMs, the organism fails to escape into the cytoplasm and its phagosome fuses to lysosomes, similar to inert particles.
Collapse
Affiliation(s)
- Marina Santic
- Department of Microbiology and Immunology, Room 316, University of Louisville College of Medicine, 319 Abraham Flexner Way 55A, Louisville, KY 40202, USA
| | | | | |
Collapse
|
32
|
Ness TL, Carpenter KJ, Ewing JL, Gerard CJ, Hogaboam CM, Kunkel SL. CCR1 and CC chemokine ligand 5 interactions exacerbate innate immune responses during sepsis. THE JOURNAL OF IMMUNOLOGY 2005; 173:6938-48. [PMID: 15557190 DOI: 10.4049/jimmunol.173.11.6938] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CCR1 has previously been shown to play important roles in leukocyte trafficking, pathogen clearance, and the type 1/type 2 cytokine balance, although very little is known about its role in the host response during sepsis. In a cecal ligation and puncture model of septic peritonitis, CCR1-deficient (CCR1(-/-)) mice were significantly protected from the lethal effects of sepsis when compared with wild-type (WT) controls. The peritoneal and systemic cytokine profile in CCR1(-/-) mice was characterized by a robust, but short-lived and regulated antibacterial response. CCR1 expression was not required for leukocyte recruitment, suggesting critical differences extant in the activation of WT and CCR1(-/-) resident or recruited peritoneal cells during sepsis. Peritoneal macrophages isolated from naive CCR1(-/-) mice clearly demonstrated enhanced cytokine/chemokine generation and antibacterial responses compared with similarly treated WT macrophages. CCR1 and CCL5 interactions markedly altered the inflammatory response in vivo and in vitro. Administration of CCL5 increased sepsis-induced lethality in WT mice, whereas neutralization of CCL5 improved survival. CCL5 acted in a CCR1-dependent manner to augment production of IFN-gamma and MIP-2 to damaging levels. These data illustrate that the interaction between CCR1 and CCL5 modulates the innate immune response during sepsis, and both represent potential targets for therapeutic intervention.
Collapse
MESH Headings
- Animals
- Ascitic Fluid/cytology
- Ascitic Fluid/immunology
- Ascitic Fluid/pathology
- Cecum
- Chemokine CCL5/biosynthesis
- Chemokine CCL5/deficiency
- Chemokine CCL5/genetics
- Chemokine CCL5/physiology
- Chemokines/biosynthesis
- Chemotaxis, Leukocyte/genetics
- Chemotaxis, Leukocyte/immunology
- Colony Count, Microbial
- Cytokines/biosynthesis
- Female
- Genetic Predisposition to Disease
- Immunity, Innate
- Ligation
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- NF-kappa B/metabolism
- Peritonitis/genetics
- Peritonitis/immunology
- Peritonitis/microbiology
- Peritonitis/mortality
- Punctures
- Receptors, CCR1
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/physiology
- Sepsis/genetics
- Sepsis/immunology
- Sepsis/microbiology
- Sepsis/mortality
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Traci L Ness
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
33
|
Liu D, Liu XY, Robinson D, Burnett C, Jackson C, Seele L, Veach RA, Downs S, Collins RD, Ballard DW, Hawiger J. Suppression of Staphylococcal Enterotoxin B-induced Toxicity by a Nuclear Import Inhibitor. J Biol Chem 2004; 279:19239-46. [PMID: 14732709 DOI: 10.1074/jbc.m313442200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Staphylococcal enterotoxin B and related toxins that target T cells have the capacity to elicit systemic inflammation, tissue injury, and death. Genes that encode mediators of inflammation can be globally inhibited by blocking the nuclear import of stress-responsive transcription factors. Here we show that cell-permeant peptides targeting Rch1/importin alpha/karyopherin alpha 2, a nuclear import adaptor protein, are delivered to T cells where they inhibit the staphylococcal enterotoxin B-induced production of inflammatory cytokines ex vivo in cultured primary spleen cells and in vivo. The systemic production of tumor necrosis factor alpha, interferon gamma, and interleukin-6 was attenuated in mice either by a cell-permeant cyclized form of SN50 peptide or by a transgene whose product suppresses the nuclear import of transcription factor nuclear factor kappa B in T cells. The extent of liver apoptosis and hemorrhagic necrosis was also reduced, which correlated with significantly decreased mortality rates. These findings highlight nuclear import inhibitors as a potentially useful countermeasure for staphylococcal enterotoxin B and other toxins that trigger harmful systemic inflammatory responses.
Collapse
Affiliation(s)
- Danya Liu
- Departments of Microbiology and Immunology and Pathology, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Cowley SC, Elkins KL. Multiple T cell subsets control Francisella tularensis LVS intracellular growth without stimulation through macrophage interferon gamma receptors. J Exp Med 2003; 198:379-89. [PMID: 12885873 PMCID: PMC2194083 DOI: 10.1084/jem.20030687] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2003] [Accepted: 05/23/2003] [Indexed: 11/21/2022] Open
Abstract
A variety of data suggest that in vivo production of interferon (IFN)-gamma is necessary, but not sufficient, for expression of secondary protective immunity against intracellular pathogens. To discover specific IFN-gamma-independent T cell mediated mechanisms, we took advantage of an in vitro culture system that models in vivo immune responses to the intracellular bacterium Francisella tularensis live vaccine strain (LVS). LVS-immune lymphocytes specifically controlled 99% of the growth of LVS in wild-type murine bone marrow-derived macrophages. Surprisingly, LVS-immune lymphocytes also inhibited LVS intracellular growth by as much as 95% in macrophages derived from IFN-gamma receptor knockout (IFNgammaR KO) mice. CD8+ T cells, and to a lesser degree CD4+ T cells, controlled LVS intracellular growth in both wild-type and IFNgammaR KO macrophages. Further, a unique population of Thy1+alphabeta+CD4-CD8- cells that was previously suggested to operate during secondary immunity to LVS in vivo strongly controlled LVS intracellular growth in vitro. A large proportion of the inhibition of LVS intracellular growth in IFNgammaR KO macrophages by all three T cell subsets could be attributed to tumor necrosis factor (TNF) alpha. Thus, T cell mechanisms exist that control LVS intracellular growth without acting through the IFN-gamma receptor; such control is due in large part to TNF-alpha, and is partially mediated by a unique double negative T cell subpopulation.
Collapse
Affiliation(s)
- Siobhán C Cowley
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, MD 20852, USA.
| | | |
Collapse
|
35
|
Kato C, Mikami M, Saito K. Nitric oxide production and iNOS mRNA expression in mice induced by repeated stimulation with live Fusobacterium nucleatum. Microbiol Immunol 2001; 45:69-78. [PMID: 11270609 DOI: 10.1111/j.1348-0421.2001.tb01260.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There have been few studies on the detection of direct nitric oxide (NO) production and interferon-gamma (IFN-gamma) in vivo without using animal cell culture. We questioned whether NO and IFN-gamma could be produced at the site of infection. The peritoneal cavity of mice was used as the local infection model. NO and IFN-gamma in abdominal washings from these mice were measured directly at various times after injection of Fusobacterium nucleatum, a gram-negative rod periodontal pathogen. The mice were divided into three groups: those treated with live bacteria (LB), those treated with heat-killed bacteria (HKB) and those untreated: normal (N). These mice were compared on the basis of cell filtration, NO and IFN-gamma production by injection of live bacteria (LFn) or heat-killed bacteria (HKFn). In the LB group, the total cell number increased corresponding to an increase in neutrophils after injection of both LFn and HKFn. A low level of NO was constantly produced in abdominal washings, but a significant amount of NO was synthesized in the LB group only 12 hr to 24 hr after injection of LFn. At the same time iNOS enzyme activity and iNOS mRNA expression were detected. IFN-gamma, which may contribute to enhance NO production, was also secreted at a high level from peritoneal exudate cells (PEC) at 12 hr and 24 hr in the LB group by stimulation of LFn. At 12 hr and 24 hr, iNOS positive cells in the LB group by infection of LFn were identified and shown to contain mostly macrophages. These findings indicate that live bacteria play important roles in NO production by macrophages. It is suggested that NO may contribute to the inflammatory response during F. nucleatum infection in periodontitis.
Collapse
Affiliation(s)
- C Kato
- Department of Oral Microbiology, School of Dentistry, Nippon Dental University at Niigata, Niigata, Japan.
| | | | | |
Collapse
|
36
|
Neumann NF, Stafford JL, Barreda D, Ainsworth AJ, Belosevic M. Antimicrobial mechanisms of fish phagocytes and their role in host defense. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2001; 25:807-825. [PMID: 11602197 DOI: 10.1016/s0145-305x(01)00037-4] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Phagocytosis is a primitive defense mechanism in all multicellular animals. Phagocytes such as macrophages and neutrophils play an important role in limiting the dissemination of infectious agents, and are responsible for the eventual destruction of phagocytosed pathogens. These cells have evolved elaborate killing mechanisms for destroying pathogens. In addition to their repertoire of degradative enzymes and antimicrobial peptides, macrophages and neutrophils can be activated to produce a number of highly toxic molecules. Production of reactive oxygen and nitrogen intermediates by these cells are potent cytotoxic mechanisms against bacteria and protozoan pathogens. Studies in fish suggest that the biological basis of these inducible killing mechanisms is similar to those described in mammals. More recent work suggest novel roles for regulating these killing responses in fish. In this review, we describe the biological basis of these killing mechanisms and how they are regulated in fish.
Collapse
Affiliation(s)
- N F Neumann
- National Water Research Institute, Canada Center for Inland Waters, Burlington, Ontario, Canada L7R 4A6.
| | | | | | | | | |
Collapse
|
37
|
Green SJ, Aniagolu J, Raney JJ. Oxidative Metabolism of Murine Macrophages. ACTA ACUST UNITED AC 2001; Chapter 14:Unit 14.5. [DOI: 10.1002/0471142735.im1405s12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
38
|
Byrom B, Obwolo M, Barbet AF, Mahan SM. A polarized Th1 type immune response to Cowdria ruminantium infection is detected in immune DBA/2 mice. J Parasitol 2000; 86:983-92. [PMID: 11128522 DOI: 10.1645/0022-3395(2000)086[0983:apttir]2.0.co;2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Immune responses to Cowdria ruminantium, an intracellular organism that causes heartwater in domestic ruminants, were characterized in a DBA/2 mouse model. Immunity induced by infection and treatment was adoptively transferable by splenocytes and could be abrogated by in vivo depletion of T cells but not by inhibition of nitric oxide synthase using NG-monomethyl-L-arginine. IgG2a and IgG2b C. ruminantium-specific responses were detected in immune mice. Culture supernatants of splenocytes from immune DBA/2 mice, which were stimulated with crude C. ruminantium antigens or recombinant major antigenic proteins 1 or 2, contained significant levels of interferon (IFN)-gamma and interleukin (IL)-6, but insignificant levels of IL-1alpha, IL-2, IL-4, IL-5, IL-10, IL-12, tumor necrosis factor-alpha (TNF), and nitric oxide. A similar response was detected during primary infection, although IFN-gamma levels decreased significantly during clinical illness and then increased following natural or antibiotic-aided recovery. These data support the conclusion that protective immunity to C. ruminantium in DBA/2 mice is mediated by T cells and is associated with a polarized T helper 1 type of immune response. This murine model could be utilized to screen for protective C. ruminantium antigens that provoke Th1 type immune responses and for evaluation of these antigens in recombinant vaccines against heartwater.
Collapse
Affiliation(s)
- B Byrom
- UF/USAID/SADC Heartwater Research Project, Central Veterinary Diagnostic and Research Laboratory, Harare, Zimbabwe
| | | | | | | |
Collapse
|
39
|
Cho KJ, Han SH, Kim BY, Hwang SG, Park KK, Yang KH, Chung AS. Chlorophyllin suppression of lipopolysaccharide-induced nitric oxide production in RAW 264.7 cells. Toxicol Appl Pharmacol 2000; 166:120-7. [PMID: 10896853 DOI: 10.1006/taap.2000.8958] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chlorophyllin (CHL), a water-soluble derivative of chlorophyll, functions as an anticarcinogen and antioxidant. In the present study, we investigated the effect of CHL on nitric oxide production in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. Treatment with CHL inhibited nitric oxide production in the LPS-stimulated RAW 264. 7 cells in a dose-related manner. Competitive RT-PCR analysis, using a DNA competitor as an internal standard, demonstrated that the treatment with 1, 10, and 50 microM CHL decreased LPS-induced iNOS mRNA expression in a concentration-dependent manner. Since the expression of the iNOS gene is mainly regulated by NF-kappaB, we then examined the effects of CHL on the NF-kappaB DNA binding activity, using an electrophoretic mobility shift assay. CHL down-regulated the NF-kappaB DNA binding on its cognate recognition site at the concentrations just noted. Employing a transfection and reporter gene expression system with p(NF-kappaB)(3)-chloramphenicol acetyl transferase (CAT), the treatment of CHL produced a dose-dependent inhibition of CAT activity in RAW 264.7 cells. Furthermore, CHL partially restored LPS-decreased IkappaBalpha, an inhibitory protein against NF-kappaB activation, in the cytosolic extract from the LPS-treated cells determined by immunoblot analysis. CHL also protected the hydroxyl radical-induced cytotoxicity in RAW 264.7 cells, indicating its antioxidant effect. These results suggest that CHL suppresses the nitric oxide production and iNOS mRNA expression mediated by the inhibition of NF-kappaB activation, and its action mechanism may be based on its antioxidant effect.
Collapse
Affiliation(s)
- K J Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 373-1, Kusong-dong, Yusong-gu, Taejon, 305-701, Korea
| | | | | | | | | | | | | |
Collapse
|
40
|
Currier AR, Ziegler MH, Riley MM, Babcock TA, Telbis VP, Carlin JM. Tumor necrosis factor-alpha and lipopolysaccharide enhance interferon-induced antichlamydial indoleamine dioxygenase activity independently. J Interferon Cytokine Res 2000; 20:369-76. [PMID: 10805371 DOI: 10.1089/107999000312306] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In macrophages, interleukin-1 (IL-1) and lipopolysaccharide (LPS) enhance the antichlamydial effect of interferon-gamma (IFN-gamma) by increasing indoleamine 2,3-dioxygenase (IDO) activity in a dose-dependent manner. Our objectives were to characterize the antichlamydial effect of tumor necrosis factor-alpha (TNF-alpha) on IFN-induced IDO activity and to establish the relationship between LPS and TNF-alpha in IDO potentiation. TNF-alpha inhibited chlamydial growth in a dose-dependent manner only in IFN-treated macrophages. Furthermore, excess tryptophan reversed the effect of combined cytokine treatment, indicating that IDO alone was responsible for chlamydial inhibition. The promonocyte THP-1 cell line, previously used to model the effect of IL-1 on IDO mRNA expression, was treated with IFN-gamma and increasing concentrations of LPS or TNF-alpha. IDO mRNA was quantified by RT-PCR, and IDO activity was measured by HPLC at 24 and 48 h after treatment, respectively. Both LPS and TNF-alpha enhanced IDO activity and IDO mRNA expression, with maximal IDO induction at 100 ng/ml LPS or 5 ng/ml TNF-alpha. Anti-TNF-alpha failed to neutralize the effects of LPS treatment, and insufficient TNF-alpha or IL-1 was produced by LPS-treated THP-1 cells to account for the enhancing effect of LPS, indicating that the effect of LPS on IDO was independent of TNF-alpha and IL-1.
Collapse
Affiliation(s)
- A R Currier
- Department of Microbiology, Miami University, Oxford, OH 45056, USA
| | | | | | | | | | | |
Collapse
|
41
|
Petricevich VL, Alves RC. Role of cytokines and nitric oxide in the induction of tuberculostatic macrophage functions. Mediators Inflamm 2000; 9:261-9. [PMID: 11213909 PMCID: PMC1781774 DOI: 10.1080/09629350020027564] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
The aim of this study was to determine phenotypic differences when BCG invades macrophages. Bacilli prepared from the same BCG primary seed, but produced in different culture media, were analysed with respect to the ability to stimulate macrophages and the susceptibility to treatment with cytokines and nitric oxide (NO). Tumour necrosis factor (TNF) activity was assayed by measuring its cytotoxic activity on L-929 cells, interleukin-6 (IL-6) and interferon-gamma (IFN-gamma) were assayed by enzyme-linked immunosorbent assay (ELISA), whereas NO levels were detected by Griess colorimetric reactions in the culture supernatant of macrophages incubated with IFN-gamma, TNF or NO and subsequently exposed to either BCG-I or BCG-S. We found that BCG-I and BCG-S bacilli showed different ability to simulate peritoneal macrophages. Similar levels of IL-6 were detected in stimulated macrophages with lysate from two BCG samples. The highest levels of TNF and IFN-gamma were observed in macrophages treated with BCG-S and BCG-I, respectively. The highest levels of NO were observed in cultures stimulated for 48 h with BCG-S. We also found a different susceptibility of the bacilli to exogenous treatment with IFN-gamma and TNF which were capable of killing 60 and 70% of both bacilli, whereas NO was capable of killing about 98 and 47% of BCG-I and BCG-S, respectively. The amount of bacilli proportionally decreased with IFN-gamma and TNF, suggesting a cytokine-related cytotoxic effect. Moreover, NO also decreased the viable number of bacilli. Interestingly, NO levels of peritoneal macrophages were significantly increased after cytokine treatment. This indicates that the treatment of macrophages with cytokines markedly reduced bacilli number and presented effects on NO production. The results obtained here emphasize the importance of adequate stimulation for guaranteeing efficient killing of bacilli. In this particular case, the IFN-gamma and TNF were involved in the activation of macrophage bactericidal activity.
Collapse
Affiliation(s)
- V L Petricevich
- Laboratorio de Imunoquímica, Instituto Butantan, São Paulo, Brazil.
| | | |
Collapse
|
42
|
Dusting GJ, Akita K, Hickey H, Smith M, Gurevich V. Cyclosporin A and tacrolimus (FK506) suppress expression of inducible nitric oxide synthase in vitro by different mechanisms. Br J Pharmacol 1999; 128:337-44. [PMID: 10510443 PMCID: PMC1571634 DOI: 10.1038/sj.bjp.0702782] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/1999] [Revised: 06/16/1999] [Accepted: 06/18/1999] [Indexed: 11/09/2022] Open
Abstract
1 The effects of the immunosuppressant drugs cyclosporin A and tacrolimus (FK506) on nitric oxide synthesis were examined in a murine macrophage cell line (J774) and rat vascular smooth muscle cells (VSMC) in culture for 24 and 48 h, respectively. 2 Cyclosporin A (0.01-10 microM) inhibited by up to 90% accumulation of nitrite induced by lipopolysaccharide (LPS) in both cell lines, but FK506 (0.01-10 microM) had a weaker effect on nitrite accumulation in these cells. Cyclosporin A and FK506 (at 1 microM) also significantly inhibited nitrite production induced by recombinant murine interferon-gamma (rIFNgamma) and recombinant murine interleukin-1beta (rIL-1beta) in J774 and VSMC, respectively. 3 In J774 cells, cyclosporin A (but not FK506) at 1 microM was inhibitory when co-incubated with the inducing agents but not when the cells were treated with the immunosuppressant before or after the inducer. In VSMC, nitrite production was inhibited by co-incubation of cyclosporin A or FK506 with the inducer, or when the immunosuppressants were pre-incubated with cells. In contrast, N-monomethyl L-arginine (NMMA) abolished nitrite production when incubated with either cell type during or after addition of inducing agent, but not if cells were preincubated with NMMA. 4 RNA extracted from treated J774 and VSMC was subjected to reverse transcription-polymerase chain reaction (RT-PCR). Cyclosporin A, but not FK506, suppressed expression of mRNA for NOS2 in a concentration-dependent manner when co-incubated with LPS. 5 The fact that the potency difference between cyclosporin A and FK506 for NO suppression is the opposite to that for inhibition of interleukin-2 generation suggests that the immunosuppressants act in J774 macrophages and VSMC through intracellular mechanisms that differ from those elucidated in T-cells. Cyclosporin A suppresses NOS2 gene transcription, but FK506 acts post-transcriptionally to suppress NO generation in VSMC. 6 Taken together the present data suggest that therapeutic concentrations of cyclosporin A, but not FK506, might well suppress NO production, but FK506 would not have this effect. Suppression of NO might contribute to the side effects of hypertension and nephrotoxicity associated with long-term use of cyclosporin A to prevent transplant rejection.
Collapse
Affiliation(s)
- G J Dusting
- Howard Florey Institute, The University of Melbourne, Parkville, Victoria 3052, Australia
| | | | | | | | | |
Collapse
|
43
|
Fang L, Nowicki BJ, Dong YL, Yallampalli C. Localized increase in nitric oxide production and the expression of nitric oxide synthase isoforms in rat uterus with experimental intrauterine infection. Am J Obstet Gynecol 1999; 181:601-9. [PMID: 10486470 DOI: 10.1016/s0002-9378(99)70499-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE We recently reported that nitric oxide was associated with increased mortality among pregnant rats with intrauterine infection. In our current study we investigated the expression of different isoforms of nitric oxide synthases and nitric oxide in the nonpregnant rat uterus with experimental intrauterine infection. STUDY DESIGN Pathogenic Escherichia coli was inoculated into the uterine lumen of ovariectomized rats. Animals were killed after inoculation, and uterine horns were collected for assessing nitric oxide production with high-performance liquid chromatography and nitric oxide synthase (type II and type III) protein expression with Western immunoblotting and immunofluorescence methods. RESULTS (1) Nitric oxide production increased in the infected uterine horn in a time-dependent manner after intrauterine infection but did not increase in the uninfected horn. (2) Nitric oxide synthase type III protein contents did not show a difference between infected and uninfected horns, and type III nitric oxide synthase was expressed by the epithelial cells and smooth muscle cells. (3) Type II nitric oxide synthase was abundantly expressed in infected horns but was not expressed in uninfected horns. Immunofluorescence data indicated that macrophages and natural killer cells, located in the endometrial layer clustering around epithelial cells, expressed type II protein. CONCLUSION We suggest that localized increase in type II nitric oxide synthase expression and nitric oxide production occurs in response to intrauterine infection and that the nitric oxide system may play a role in host response to restrict the infection.
Collapse
Affiliation(s)
- L Fang
- Department of Obstetrics, The University of Texas Medical Branch, Galveston, USA
| | | | | | | |
Collapse
|
44
|
Haque A, Graille M, Kasper LH, Haque S. Immunization with heat-killed Toxoplasma gondii stimulates an early IFN-gamma response and induces protection against virulent murine malaria. Vaccine 1999; 17:2604-11. [PMID: 10418909 DOI: 10.1016/s0264-410x(99)00050-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In this study, we describe protection of BALB/c mice by immunization with heat-killed T. gondii tachyzoites against infection with Plasmodium yoelii 17XL which causes cerebral malaria and death in mice by day 7-8 post infection. Immunization resulted significant reduction in parasitemia at the peak period of infection. Protection induced by heat-killed T. gondii was associated with marked increase in NK cell number and IFN-gamma mRNA expression early in the infection. The level of IFN-gamma or TNF-alpha was found to diminish in T. gondii-treated mice as the infection progressed to the late stage. This declined response of IFN-gamma or TNF-alpha was associated with marked increase in the expression of IL-10, a counterregulatory cytokine. Pretreatment of mice with live T. gondii induced poor level of protection as compared with that of heat-killed parasites. Mice that received P. yoelii infection alone, had an elevated IFN-gamma response in the late stage of infection. Development of cerebral malaria in untreated mice was accompanied by an augmented production of TNF-alpha and nitric oxide (NO), the proinflammatory mediators. These findings suggest that nonspecific immunization with T. gondii leads to restoration of an early IFN-gamma response in P. yoelii-infected mice and in the establishment of an immunoregulatory mechanism that effectively antagonizes the disease-promoting effects of proinflammatory cytokines in the late phase of infection.
Collapse
Affiliation(s)
- A Haque
- Immunologie et Génétique des Maladies Parasitaires, INSERM U399, Faculté de Medecine, La Timone, Marseille, France.
| | | | | | | |
Collapse
|
45
|
Hirunpetcharat C, Finkelman F, Clark IA, Good MF. Malaria parasite-specific Th1-like T cells simultaneously reduce parasitemia and promote disease. Parasite Immunol 1999; 21:319-29. [PMID: 10354354 DOI: 10.1046/j.1365-3024.1999.00234.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
CD4+ T cells have been implicated in immunity to the blood stages of malaria and cytokines associated with both monocyte and T cell activation have been implicated in disease. To determine whether specific T cells capable of inhibiting parasite growth can also mediate pathology we have transfused populations of Plasmodium berghei-specific T cells into normal and immunodeficient naive mice. We observed that they could inhibit parasite growth but were unable to save the animals which exhibited significantly greater anaemia and weight loss than control infected animals receiving either no T cells or T cells specific for ovalbumin. T cell-dependent tomour necrosis factor (TNF)alpha was a critical component in both parasite killing and disease promotion. Experiments with blocking antibodies demonstrated that all T-cell mediated antiparasitic immunity and all T-cell mediated weight loss was TNF-dependent. Blocking TNF-alpha in mice that received parasite-specific T cells prolonged the survival of the mice. Nitric oxide demonstrated no antiparasite effect, but was involved in the regulation of T-cell mediated weight loss. The data thus show that while parasite-specific CD4+ T cells can significantly limit parasite growth, such an effect need not be beneficial to the host, and that TNF-alpha and nitric oxide are critical effector molecules operating downstream of parasite-specific T cells in both immunity and disease.
Collapse
Affiliation(s)
- C Hirunpetcharat
- The Queensland Institute of Medical Research, Royal Brisbane Hospital, Brisbane 4029, Australia
| | | | | | | |
Collapse
|
46
|
Oliveira DM, Silva-Teixeira DN, Carmo SA, Goes AM. Role of nitric oxide on human schistosomiasis mansoni: upregulation of in vitro granuloma formation by N omega-nitro-L-arginine methyl ester. Nitric Oxide 1998; 2:57-65. [PMID: 9706743 DOI: 10.1006/niox.1997.0164] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Over the past decade, nitric oxide has been intensely studied due to its relevance as a widespread intra- and intercellular messenger and as a cytotoxin released during several physiopathological events, including immunological reactions and inflammation. In the present paper, we investigate the effect of inhibition of NO synthesis, using an analogue of L-arginine, N omega-nitro-L-arginine methyl ester (L-NAME), on in vitro granulomatous formation of human peripheral blood mononuclear cells (PBMC) from Schistosoma mansoni-infected individuals. The results demonstrated that human PBMC are capable of in vitro NO production and that inhibition of its production through the addition of L-NAME is responsible for exacerbating granulomatous reaction. This L-NAME-induced granuloma enhancement (ranging from 30 to 65%) was measured using the granuloma index. Furthermore, we observed a general time-dependent increase in NO production during the period of cell culture (21 days) and an inverse relationship between nitrite detection and granuloma reactivity. Collectively, our results point to a possible regulatory role of NO on the development of granulomatous inflammation.
Collapse
Affiliation(s)
- D M Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | | | | |
Collapse
|
47
|
Everest P, Roberts M, Dougan G. Susceptibility to Salmonella typhimurium infection and effectiveness of vaccination in mice deficient in the tumor necrosis factor alpha p55 receptor. Infect Immun 1998; 66:3355-64. [PMID: 9632605 PMCID: PMC108352 DOI: 10.1128/iai.66.7.3355-3364.1998] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mice defective in the ability to produce the tumor necrosis factor alpha p55 receptor (TNFalphap55R) were orally challenged with a number of Salmonella typhimurium HWSH derivatives that differ in virulence. In comparison to TNFalphap55R+/+ mice, TNFalphap55R-/- mice succumbed earlier to challenge with wild-type S. typhimurium HWSH and S. typhimurium HWSH purE. In contrast, TNFalphap55R-/- mice were able to control an S. typhimurium HWSH aroA challenge, although greater numbers of Salmonella organisms were present in the tissues for a longer time period than was observed with TNFalphap55R+/+ mice. Vaccination of normal and TNFalphap55R knockout animals with S. typhimurium HWSH aroA showed that TNFalphap55R-/- mice, unlike TNFalphap55R+/+ mice, were not protected against a virulent S. typhimurium HWSH challenge. Splenocytes from TNFalphap55R-/- mice exhibited a reduced ability to proliferate in the presence of S. typhimurium antigen compared to TNFalphap55R+/+ mice. Thus, TNFalphap55R is essential for controlling Salmonella growth in tissues and for recall of immunity in murine salmonellosis.
Collapse
Affiliation(s)
- P Everest
- Department of Biochemistry, Imperial College of Science, Technology and Medicine, London SW7 2AZ, United Kingdom
| | | | | |
Collapse
|
48
|
Cao YM, Tsuboi T, Torii M. Nitric oxide inhibits the development of Plasmodium yoelii gametocytes into gametes. Parasitol Int 1998. [DOI: 10.1016/s1383-5769(98)00014-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
49
|
Deepe GS, Seder RA. Molecular and cellular determinants of immunity to Histoplasma capsulatum. RESEARCH IN IMMUNOLOGY 1998; 149:397-406; discussion 509-10. [PMID: 9720957 DOI: 10.1016/s0923-2494(98)80763-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- G S Deepe
- Division of Infectious Diseases, University of Cincinnati College of Medicine, OH 45267-0560, USA
| | | |
Collapse
|
50
|
Sasaki S, Miura T, Nishikawa S, Yamada K, Hirasue M, Nakane A. Protective role of nitric oxide in Staphylococcus aureus infection in mice. Infect Immun 1998; 66:1017-22. [PMID: 9488390 PMCID: PMC108010 DOI: 10.1128/iai.66.3.1017-1022.1998] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This study was carried out to determine the role of nitric oxide (NO) in Staphylococcus aureus infection in mice. NO production in spleen cell cultures was induced by heat-killed S. aureus. Expression of mRNA of the inducible isoform of NO synthase (iNOS) was induced in the spleens and kidneys of S. aureus-infected mice. When mice were treated with monoclonal antibodies (MAbs) against tumor necrosis factor alpha (TNF-alpha) or gamma interferon (IFN-gamma) before S. aureus infection, the induction of iNOS mRNA expression in the kidneys was inhibited. These MAbs also inhibited NO production in spleen cell cultures stimulated with heat-killed S. aureus. NO production in the spleen cell cultures and levels of urinary nitrate plus nitrite were suppressed by treatment with aminoguanidine (AG), a selective inhibitor of iNOS. The survival rates of AG-treated mice were significantly decreased by either lethal or sublethal S. aureus infections. However, an effect of AG administration on bacterial growth was not observed in the spleens and kidneys of mice during either type of infection. Production of TNF-alpha and IFN-gamma was not affected by AG treatment in vitro and in vivo. These results suggest that NO plays an important role in protection from lethality by the infection, but the protective role of NO in host resistance against S. aureus infection was not proved. Moreover, our results show that TNF-alpha and IFN-gamma regulate NO production while NO may not be involved in the regulation of the production of these cytokines during S. aureus infection.
Collapse
Affiliation(s)
- S Sasaki
- Department of Bacteriology, School of Medicine, Hirosaki University, Japan
| | | | | | | | | | | |
Collapse
|