1
|
Bin Y, Ren J, Zhang H, Zhang T, Liu P, Xin Z, Yang H, Feng Z, Chen Z, Zhang H. Against all odds: The road to success in the development of human immune reconstitution mice. Animal Model Exp Med 2024; 7:460-470. [PMID: 38591343 PMCID: PMC11369039 DOI: 10.1002/ame2.12407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/17/2024] [Indexed: 04/10/2024] Open
Abstract
The mouse genome has a high degree of homology with the human genome, and its physiological, biochemical, and developmental regulation mechanisms are similar to those of humans; therefore, mice are widely used as experimental animals. However, it is undeniable that interspecies differences between humans and mice can lead to experimental errors. The differences in the immune system have become an important factor limiting current immunological research. The application of immunodeficient mice provides a possible solution to these problems. By transplanting human immune cells or tissues, such as peripheral blood mononuclear cells or hematopoietic stem cells, into immunodeficient mice, a human immune system can be reconstituted in the mouse body, and the engrafted immune cells can elicit human-specific immune responses. Researchers have been actively exploring the development and differentiation conditions of host recipient animals and grafts in order to achieve better immune reconstitution. Through genetic engineering methods, immunodeficient mice can be further modified to provide a favorable developmental and differentiation microenvironment for the grafts. From initially only being able to reconstruct single T lymphocyte lineages, it is now possible to reconstruct lymphoid and myeloid cells, providing important research tools for immunology-related studies. In this review, we compare the differences in immune systems of humans and mice, describe the development history of human immune reconstitution from the perspectives of immunodeficient mice and grafts, and discuss the latest advances in enhancing the efficiency of human immune cell reconstitution, aiming to provide important references for immunological related researches.
Collapse
Affiliation(s)
- Yixiao Bin
- School of Basic Medical SciencesShaanxi University of Chinese MedicineXianyangChina
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Jing Ren
- School of Basic Medical SciencesShaanxi University of Chinese MedicineXianyangChina
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Haowei Zhang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public HealthFourth Military Medical UniversityXi'anChina
| | - Tianjiao Zhang
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Peijuan Liu
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhiqian Xin
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Haijiao Yang
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhuan Feng
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhinan Chen
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Hai Zhang
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| |
Collapse
|
2
|
Zargar S, Altwaijry N, Wani TA, Alkahtani HM. Evaluation of the Possible Pathways Involved in the Protective Effects of Quercetin, Naringenin, and Rutin at the Gene, Protein and miRNA Levels Using In-Silico Multidimensional Data Analysis. Molecules 2023; 28:4904. [PMID: 37446564 DOI: 10.3390/molecules28134904] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Flavonoids are secondary metabolites that are non-essential for plant growth or survival, and they also provide numerous health benefits to humans. They are antioxidants that shield plants from the ill effects of ultraviolet light, pests, and diseases. They are beneficial to health for several reasons, including lowering inflammation, boosting cardiovascular health, and lowering cancer risk. This study looked into the physicochemical features of these substances to determine the potential pharmacological pathways involved in their protective actions. Potential targets responsible for the protective effects of quercetin, naringenin, and rutin were identified with SwissADME. The associated biological processes and protein-protein networks were analyzed by using the GeneMANIA, Metascape, and STRING servers. All the flavonoids were predicted to be orally bioavailable, with more than 90% targets as enzymes, including kinases and lyases, and with common targets such as NOS2, CASP3, CASP9, CAT, BCL2, TNF, and HMOX1. TNF was shown to be a major target in over 250 interactions. To extract the "biological meanings" from the MCODE networks' constituent parts, a GO enrichment analysis was performed on each one. The most important transcription factors in gene regulation were RELA, NFKB1, PPARG, and SP1. Treatment with quercetin, naringenin, or rutin increased the expression and interaction of the microRNAs' hsa-miR-34a-5p, hsa-miR-30b-5p, hsa-let-7a-5p, and hsa-miR-26a-1-3p. The anticancer effects of hsa-miR-34a-5p have been experimentally confirmed. It also plays a critical role in controlling other cancer-related processes such as cell proliferation, apoptosis, EMT, and metastasis. This study's findings might lead to a deeper comprehension of the mechanisms responsible for flavonoids' protective effects and could present new avenues for exploration.
Collapse
Affiliation(s)
- Seema Zargar
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Nojood Altwaijry
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Tanveer A Wani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
3
|
Zhang K, Sowers ML, Cherryhomes EI, Singh VK, Mishra A, Restrepo BI, Khan A, Jagannath C. Sirtuin-dependent metabolic and epigenetic regulation of macrophages during tuberculosis. Front Immunol 2023; 14:1121495. [PMID: 36993975 PMCID: PMC10040548 DOI: 10.3389/fimmu.2023.1121495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/01/2023] [Indexed: 03/14/2023] Open
Abstract
Macrophages are the preeminent phagocytic cells which control multiple infections. Tuberculosis a leading cause of death in mankind and the causative organism Mycobacterium tuberculosis (MTB) infects and persists in macrophages. Macrophages use reactive oxygen and nitrogen species (ROS/RNS) and autophagy to kill and degrade microbes including MTB. Glucose metabolism regulates the macrophage-mediated antimicrobial mechanisms. Whereas glucose is essential for the growth of cells in immune cells, glucose metabolism and its downsteam metabolic pathways generate key mediators which are essential co-substrates for post-translational modifications of histone proteins, which in turn, epigenetically regulate gene expression. Herein, we describe the role of sirtuins which are NAD+-dependent histone histone/protein deacetylases during the epigenetic regulation of autophagy, the production of ROS/RNS, acetyl-CoA, NAD+, and S-adenosine methionine (SAM), and illustrate the cross-talk between immunometabolism and epigenetics on macrophage activation. We highlight sirtuins as emerging therapeutic targets for modifying immunometabolism to alter macrophage phenotype and antimicrobial function.
Collapse
Affiliation(s)
- Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Mark L. Sowers
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ellie I. Cherryhomes
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Vipul K. Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Blanca I. Restrepo
- University of Texas Health Houston, School of Public Health, Brownsville, TX, United States
| | - Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| |
Collapse
|
4
|
Rasaei R, Tyagi A, Rasaei S, Lee SJ, Yang SR, Kim KS, Ramakrishna S, Hong SH. Human pluripotent stem cell-derived macrophages and macrophage-derived exosomes: therapeutic potential in pulmonary fibrosis. Stem Cell Res Ther 2022; 13:433. [PMID: 36056418 PMCID: PMC9438152 DOI: 10.1186/s13287-022-03136-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 08/14/2022] [Indexed: 11/10/2022] Open
Abstract
Pulmonary fibrosis (PF) is a fatal chronic disease characterized by accumulation of extracellular matrix and thickening of the alveolar wall, ultimately leading to respiratory failure. PF is thought to be initiated by the dysfunction and aberrant activation of a variety of cell types in the lung. In particular, several studies have demonstrated that macrophages play a pivotal role in the development and progression of PF through secretion of inflammatory cytokines, growth factors, and chemokines, suggesting that they could be an alternative therapeutic source as well as therapeutic target for PF. In this review, we describe the characteristics, functions, and origins of subsets of macrophages involved in PF and summarize current data on the generation and therapeutic application of macrophages derived from pluripotent stem cells for the treatment of fibrotic diseases. Additionally, we discuss the use of macrophage-derived exosomes to repair fibrotic lung tissue.
Collapse
Affiliation(s)
- Roya Rasaei
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehakgil, Chuncheon, Gangwon-do, 24431, South Korea
| | - Apoorvi Tyagi
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Shima Rasaei
- Department of Cellular and Molecular Science, Falavarjan Branch, Islamic Azad University, Falavarjan, Iran
| | - Seung-Joon Lee
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehakgil, Chuncheon, Gangwon-do, 24431, South Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea.
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehakgil, Chuncheon, Gangwon-do, 24431, South Korea.
- Institute of Medical Science, Kangwon National University, Chuncheon, 24341, South Korea.
- KW-Bio Co., Ltd, Wonju, South Korea.
| |
Collapse
|
5
|
Current Status, Barriers, and Future Directions for Humanized Mouse Models to Evaluate Stem Cell–Based Islet Cell Transplant. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1387:89-106. [DOI: 10.1007/5584_2022_711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
6
|
Ritter M, Bresgen N, Kerschbaum HH. From Pinocytosis to Methuosis-Fluid Consumption as a Risk Factor for Cell Death. Front Cell Dev Biol 2021; 9:651982. [PMID: 34249909 PMCID: PMC8261248 DOI: 10.3389/fcell.2021.651982] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
The volumes of a cell [cell volume (CV)] and its organelles are adjusted by osmoregulatory processes. During pinocytosis, extracellular fluid volume equivalent to its CV is incorporated within an hour and membrane area equivalent to the cell's surface within 30 min. Since neither fluid uptake nor membrane consumption leads to swelling or shrinkage, cells must be equipped with potent volume regulatory mechanisms. Normally, cells respond to outwardly or inwardly directed osmotic gradients by a volume decrease and increase, respectively, i.e., they shrink or swell but then try to recover their CV. However, when a cell death (CD) pathway is triggered, CV persistently decreases in isotonic conditions in apoptosis and it increases in necrosis. One type of CD associated with cell swelling is due to a dysfunctional pinocytosis. Methuosis, a non-apoptotic CD phenotype, occurs when cells accumulate too much fluid by macropinocytosis. In contrast to functional pinocytosis, in methuosis, macropinosomes neither recycle nor fuse with lysosomes but with each other to form giant vacuoles, which finally cause rupture of the plasma membrane (PM). Understanding methuosis longs for the understanding of the ionic mechanisms of cell volume regulation (CVR) and vesicular volume regulation (VVR). In nascent macropinosomes, ion channels and transporters are derived from the PM. Along trafficking from the PM to the perinuclear area, the equipment of channels and transporters of the vesicle membrane changes by retrieval, addition, and recycling from and back to the PM, causing profound changes in vesicular ion concentrations, acidification, and-most importantly-shrinkage of the macropinosome, which is indispensable for its proper targeting and cargo processing. In this review, we discuss ion and water transport mechanisms with respect to CVR and VVR and with special emphasis on pinocytosis and methuosis. We describe various aspects of the complex mutual interplay between extracellular and intracellular ions and ion gradients, the PM and vesicular membrane, phosphoinositides, monomeric G proteins and their targets, as well as the submembranous cytoskeleton. Our aim is to highlight important cellular mechanisms, components, and processes that may lead to methuotic CD upon their derangement.
Collapse
Affiliation(s)
- Markus Ritter
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria
- Institute for Physiology and Pathophysiology, Paracelsus Medical University, Nuremberg, Germany
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis und Rehabilitation, Salzburg, Austria
- Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
| | - Nikolaus Bresgen
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | | |
Collapse
|
7
|
Lyadova I, Gerasimova T, Nenasheva T. Macrophages Derived From Human Induced Pluripotent Stem Cells: The Diversity of Protocols, Future Prospects, and Outstanding Questions. Front Cell Dev Biol 2021; 9:640703. [PMID: 34150747 PMCID: PMC8207294 DOI: 10.3389/fcell.2021.640703] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/25/2021] [Indexed: 12/23/2022] Open
Abstract
Macrophages (Mφ) derived from induced pluripotent stem cells (iMphs) represent a novel and promising model for studying human Mφ function and differentiation and developing new therapeutic strategies based on or oriented at Mφs. iMphs have several advantages over the traditionally used human Mφ models, such as immortalized cell lines and monocyte-derived Mφs. The advantages include the possibility of obtaining genetically identical and editable cells in a potentially scalable way. Various applications of iMphs are being developed, and their number is rapidly growing. However, the protocols of iMph differentiation that are currently used vary substantially, which may lead to differences in iMph differentiation trajectories and properties. Standardization of the protocols and identification of minimum required conditions that would allow obtaining iMphs in a large-scale, inexpensive, and clinically suitable mode are needed for future iMph applications. As a first step in this direction, the current review discusses the fundamental basis for the generation of human iMphs, performs a detailed analysis of the generalities and the differences between iMph differentiation protocols currently employed, and discusses the prospects of iMph applications.
Collapse
Affiliation(s)
- Irina Lyadova
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | | | | |
Collapse
|
8
|
Yoon SY, Sa SJ, Cho ES, Ko HS, Choi JW, Kim JS. Effects of Zinc Oxide and Arginine on the Intestinal Microbiota and Immune Status of Weaned Pigs Subjected to High Ambient Temperature. Animals (Basel) 2020; 10:ani10091537. [PMID: 32878254 PMCID: PMC7552118 DOI: 10.3390/ani10091537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 08/25/2020] [Indexed: 12/26/2022] Open
Abstract
This study aimed to investigate the effect of the l-arginine (Arg) inclusion and different doses of ZnO on the growth performance, intestinal microbiota and integrity, and immune status of weaned pigs. A total of 180 pigs (28-day-old) were randomly allotted to six treatments with six replicate pens in each treatment and five pigs per pen. The dietary treatments were Con (1.1% Arg); P-Zn (1.1% Arg + 2500 mg Zn as ZnO/kg diet); ARG (1.6% Arg); ZnArg1 (500 mg of Zn as ZnO/kg diet + 1.6% Arg); ZnArg2 (1000 mg of Zn as ZnO/kg diet + 1.6% Arg); ZnArg3 (2500 mg of Zn as ZnO/kg diet + 1.6% Arg). The overall result showed that the inclusion of ZnArg3 significantly improved the average daily gain of pigs compared with the Con treatment. There was a reduction in feed intake in pigs fed the Con diet compared with pigs fed the ZnArg3 diet at phase 1 and overall. At phase 1, pigs fed the ZnArg3 diet and P-Zn diet showed a decreased population of Clostridium spp. in the ileum compared with those of the Con treatment. In addition, a lower ileal Clostridium spp. population was detected in pigs fed the ZnArg2 diet compared with pigs fed the Con diet. The pigs fed ZnArg1 and ZnArg3 diets showed a greater villus height of duodenum compared with the Con and P-Zn treatments. The pigs in the Con treatment showed increased mRNA expression of heat shock protein-27 in the liver compared with the P-Zn, ZnArg1, ZnArg2, and ZnArg3 treatments. When fed the basal diet, mRNA expressions of interleukin-6 were increased in the muscle compared with the ZnArg3 treatment. Dietary supplementation with ZnArg2 decreased the mRNA expressions of interferon-γ in the muscle compared with the Con treatment. Supplementation with P-Zn, ZnArg1, ZnArg2, and ZnArg3 decreased mRNA expressions of tumor necrosis factor-α (TNF-α) compared with the Con treatment. The mRNA gene expressions of interleukin-4 were decreased in the jejunum of pigs fed P-Zn, ARG, ZnArg1, ZnArg2, and ZnArg3 diets compared with pigs fed the Con diet. The jejunum gene expression of toll-like receptor-4 was upregulated in the Con and ARG treatments compared with the ZnArg1 and ZnArg3. The ZnArg1, ZnArg2, and ZnArg3 treatments showed lower mRNA expression of TNF-α compared with the Con treatment. In conclusion, there was no difference in growth performance, intestinal microbiota, gene expression of interleukins between ZnArg1 and ZnArg3 treatments. Therefore, the low level of ZnO (500 mg/kg) plus 1.6% dietary Arg may be recommended for pigs during the weaning stress.
Collapse
Affiliation(s)
- Se Young Yoon
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea; (S.Y.Y.); (H.S.K.); (J.W.C.)
| | - Soo Jin Sa
- National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Korea; (S.J.S.); (E.S.C.)
| | - Eun Seok Cho
- National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Korea; (S.J.S.); (E.S.C.)
| | - Han Seo Ko
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea; (S.Y.Y.); (H.S.K.); (J.W.C.)
| | - Jung Woo Choi
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea; (S.Y.Y.); (H.S.K.); (J.W.C.)
| | - Jin Soo Kim
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea; (S.Y.Y.); (H.S.K.); (J.W.C.)
- Correspondence: ; Tel.: +82-33-250-8614; Fax: +82-33-259-5572
| |
Collapse
|
9
|
Pan L, Yang S, Wang J, Xu M, Wang S, Yi H. Inducible nitric oxide synthase and systemic lupus erythematosus: a systematic review and meta-analysis. BMC Immunol 2020; 21:6. [PMID: 32066371 PMCID: PMC7027241 DOI: 10.1186/s12865-020-0335-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 01/29/2020] [Indexed: 01/01/2023] Open
Abstract
Background There is a growing body of evidences indicating iNOS has involved in the pathogenesis of SLE. However, the role of iNOS in SLE is inconsistency. This systematic review was designed to evaluate the association between iNOS and SLE. Results Six studies were included, reporting on a total of 277 patients with SLE. The meta-analysis showed that SLE patients had higher expression of iNOS at mRNA level than control subjects (SMD = 2.671, 95%CI = 0.446–4.897, z = 2.35, p = 0.019), and a similar trend was noted at the protein level (SMD = 3.602, 95%CI = 1.144–6.059, z = 2.87, p = 0.004) and positive rate of iNOS (OR = 9.515, 95%CI = 1.915–47.281, z = 2.76, p = 0.006) were significantly higher in SLE group compared with control group. No significant difference was observed on serum nitrite level between SLE patients and control subjects (SMD = 2.203, 95%CI = -0.386–4.793, z = 1.64, p = 0.095). The results did not modify from different sensitivity analysis, representing the robustness of this study. No significant publication bias was detected from Egger’s test. Conclusions There was a positive correlation between increasing iNOS and SLE. However, the source of iNOS is unknown. Besides NO pathway, other pathways also should be considered. More prospective random studies are needed in order to certify our results.
Collapse
Affiliation(s)
- Lu Pan
- Central Laboratory, The First Hospital of Jilin University, Changchun, China.,Department of Pediatric Rheumatology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Sirui Yang
- Department of Pediatric Rheumatology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Jinghua Wang
- Department of Pediatric Rheumatology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Meng Xu
- Department of Pediatric Rheumatology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Shaofeng Wang
- The Institute of Epigenetic Medicine, The First Hospital of Jilin University, Changchun, China.
| | - Huanfa Yi
- Central Laboratory, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
10
|
Healy LM, Yaqubi M, Ludwin S, Antel JP. Species differences in immune-mediated CNS tissue injury and repair: A (neuro)inflammatory topic. Glia 2019; 68:811-829. [PMID: 31724770 DOI: 10.1002/glia.23746] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/04/2019] [Accepted: 10/11/2019] [Indexed: 12/13/2022]
Abstract
Cells of the adaptive and innate immune systems in the brain parenchyma and in the meningeal spaces contribute to physiologic functions and disease states in the central nervous system (CNS). Animal studies have demonstrated the involvement of immune constituents, along with major histocompatibility complex (MHC) molecules, in neural development and rare genetic disorders (e.g., colony stimulating factor 1 receptor [CSF1R] deficiency). Genome wide association studies suggest a comparable role of the immune system in humans. Although the CNS can be the target of primary autoimmune disorders, no current experimental model captures all of the features of the most common human disorder placed in this category, multiple sclerosis (MS). Such features include spontaneous onset, environmental contributions, and a recurrent/progressive disease course in a genetically predisposed host. Numerous therapeutic interventions related to antigen and cytokine specific therapies have demonstrated effectiveness in experimental autoimmune encephalomyelitis (EAE), the animal model used to define principles underlying immune-mediated mechanisms in MS. Despite the similarities in the two diseases, most treatments used to ameliorate EAE have failed to translate to the human disease. As directly demonstrated in animal models and implicated by correlative studies in humans, adaptive and innate immune constituents within the systemic compartment and resident in the CNS contribute to the disease course of neurodegenerative and neurobehavioral disorders. The expanding knowledge of the molecular properties of glial cells provides increasing insights into species related variables. These variables affect glial bidirectional interactions with the immune system as well as their own production of "immune molecules" that mediate tissue injury and repair.
Collapse
Affiliation(s)
- Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Moein Yaqubi
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Samuel Ludwin
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
11
|
Ariyaratne A, Finney CAM. Eosinophils and Macrophages within the Th2-Induced Granuloma: Balancing Killing and Healing in a Tight Space. Infect Immun 2019; 87:e00127-19. [PMID: 31285249 PMCID: PMC6759305 DOI: 10.1128/iai.00127-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Granuloma formation is a key host immune response generated to confine invading pathogens and limit extensive host damage. It consists of an accumulation of host immune cells around a pathogen. This host response has been extensively studied in the context of inflammatory diseases. However, there is much less known about Th2-type granulomas generated in response to parasitic worms. Based on in vitro data, innate immune cells within the granuloma are thought to immobilize and kill parasites but also act to repair damaged tissue. Understanding this dual function is key. The two billion people and many livestock/wild animals infected with helminths demonstrate that granulomas are not effective at clearing infection. However, the lack of high mortality highlights their importance in ensuring that parasite migration/tissue damage is restricted and wound healing is effective. In this review, we define two key cellular players (macrophages and eosinophils) and their associated molecular players involved in Th2 granuloma function. To date, the underlying mechanisms remain poorly understood, which is in part due to a lack of conclusive studies. Most have been performed in vitro rather than in vivo, using cells that have not been obtained from granulomas. Experiments using genetically modified mouse strains and/or antibody/chemical-mediated cell depletion have also generated conflicting results depending on the model. We discuss the caveats of previous studies and the new tools available that will help fill the gaps in our knowledge and allow a better understanding of the balance between immune killing and healing.
Collapse
Affiliation(s)
- Anupama Ariyaratne
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Constance A M Finney
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Reactive nitrogen species in host-bacterial interactions. Curr Opin Immunol 2019; 60:96-102. [PMID: 31200187 DOI: 10.1016/j.coi.2019.05.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/29/2019] [Accepted: 05/11/2019] [Indexed: 12/11/2022]
Abstract
Reactive nitrogen species play diverse and essential roles in host-pathogen interactions. Here, we review selected recent discoveries regarding nitric oxide (NO) in host defense and the pathogenesis of infection, mechanisms of bacterial NO resistance, production of NO by human macrophages, NO-based antimicrobial therapeutics and NO interactions with the gut microbiota.
Collapse
|
13
|
Sauter IP, Madrid KG, de Assis JB, Sá-Nunes A, Torrecilhas AC, Staquicini DI, Pasqualini R, Arap W, Cortez M. TLR9/MyD88/TRIF signaling activates host immune inhibitory CD200 in Leishmania infection. JCI Insight 2019; 4:126207. [PMID: 31092731 DOI: 10.1172/jci.insight.126207] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/11/2019] [Indexed: 02/06/2023] Open
Abstract
Virulent protozoans named Leishmania in tropical and subtropical areas produce devastating diseases by exploiting host immune responses. Amastigotes of Leishmania amazonensis stimulate macrophages to express CD200, an immunomodulatory ligand, which binds to its cognate receptor (CD200R) and inhibits the inducible nitric oxide synthase and nitric oxide (iNOS/NO) signaling pathways, thereby promoting intracellular survival. However, the mechanisms underlying CD200 induction in macrophages remain largely unknown. Here, we show that phagocytosis-mediated internalization of L. amazonensis amastigotes following activation of endosomal TLR9/MyD88/TRIF signaling is critical for inducing CD200 in infected macrophages. We also demonstrate that Leishmania microvesicles containing DNA fragments activate TLR9-dependent CD200 expression, which inhibits the iNOS/NO pathway and modulates the course of L. amazonensis infection in vivo. These findings demonstrate that Leishmania exploits TLR-signaling pathways not only to inhibit macrophage microbicidal function, but also to evade host systemic immune responses, which has many implications in the severity of the disease.
Collapse
Affiliation(s)
| | | | - Josiane B de Assis
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Anderson Sá-Nunes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana C Torrecilhas
- Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Daniela I Staquicini
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey and Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | | |
Collapse
|
14
|
Piacenza L, Trujillo M, Radi R. Reactive species and pathogen antioxidant networks during phagocytosis. J Exp Med 2019; 216:501-516. [PMID: 30792185 PMCID: PMC6400530 DOI: 10.1084/jem.20181886] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/04/2019] [Accepted: 02/04/2019] [Indexed: 11/23/2022] Open
Abstract
The generation of phagosomal cytotoxic reactive species (i.e., free radicals and oxidants) by activated macrophages and neutrophils is a crucial process for the control of intracellular pathogens. The chemical nature of these species, the reactions they are involved in, and the subsequent effects are multifaceted and depend on several host- and pathogen-derived factors that influence their production rates and catabolism inside the phagosome. Pathogens rely on an intricate and synergistic antioxidant armamentarium that ensures their own survival by detoxifying reactive species. In this review, we discuss the generation, kinetics, and toxicity of reactive species generated in phagocytes, with a focus on the response of macrophages to internalized pathogens and concentrating on Mycobacterium tuberculosis and Trypanosoma cruzi as examples of bacterial and parasitic infection, respectively. The ability of pathogens to deal with host-derived reactive species largely depends on the competence of their antioxidant networks at the onset of invasion, which in turn can tilt the balance toward pathogen survival, proliferation, and virulence over redox-dependent control of infection.
Collapse
Affiliation(s)
- Lucía Piacenza
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
15
|
Lucas AT, Robinson R, Schorzman AN, Piscitelli JA, Razo JF, Zamboni WC. Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients. Antibodies (Basel) 2019; 8:E3. [PMID: 31544809 PMCID: PMC6640706 DOI: 10.3390/antib8010003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 12/11/2022] Open
Abstract
The rapid advancement in the development of therapeutic proteins, including monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), has created a novel mechanism to selectively deliver highly potent cytotoxic agents in the treatment of cancer. These agents provide numerous benefits compared to traditional small molecule drugs, though their clinical use still requires optimization. The pharmacology of mAbs/ADCs is complex and because ADCs are comprised of multiple components, individual agent characteristics and patient variables can affect their disposition. To further improve the clinical use and rational development of these agents, it is imperative to comprehend the complex mechanisms employed by antibody-based agents in traversing numerous biological barriers and how agent/patient factors affect tumor delivery, toxicities, efficacy, and ultimately, biodistribution. This review provides an updated summary of factors known to affect the disposition of mAbs/ADCs in development and in clinical use, as well as how these factors should be considered in the selection and design of preclinical studies of ADC agents in development.
Collapse
Affiliation(s)
- Andrew T Lucas
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Ryan Robinson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Joseph A Piscitelli
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - Juan F Razo
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
16
|
López-Mejías R, Castañeda S, Genre F, Remuzgo-Martínez S, Carmona FD, Llorca J, Blanco R, Martín J, González-Gay MA. Genetics of immunoglobulin-A vasculitis (Henoch-Schönlein purpura): An updated review. Autoimmun Rev 2018; 17:301-315. [DOI: 10.1016/j.autrev.2017.11.024] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 11/16/2017] [Indexed: 12/12/2022]
|
17
|
Li G, Yamasaki R, Fang M, Masaki K, Ochi H, Matsushita T, Kira JI. Novel disease-modifying anti-rheumatic drug iguratimod suppresses chronic experimental autoimmune encephalomyelitis by down-regulating activation of macrophages/microglia through an NF-κB pathway. Sci Rep 2018; 8:1933. [PMID: 29386552 PMCID: PMC5792543 DOI: 10.1038/s41598-018-20390-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 01/17/2018] [Indexed: 12/28/2022] Open
Abstract
We aimed to elucidate the effects of iguratimod, a widely used anti-rheumatic drug with no severe side effects, on chronic experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Iguratimod was orally administered to mice immunised with myelin oligodendrocyte glycoprotein peptide 35–55. Preventive administration of iguratimod from the time of immunisation was found to markedly reduce the clinical severity of acute and chronic EAE. Pathologically, iguratimod treatment significantly reduced demyelination and infiltration of CD3+ T, F4/80+, and CD169+ cells into the spinal cord, and suppressed macrophage/microglia activation in the parenchyma at the acute and chronic stages compared with vehicle treatment. Therapeutic administration of iguratimod after the onset of clinical symptoms significantly ameliorated the clinical severity of chronic EAE and reduced demyelination, T helper (Th)1/Th17 cell infiltration, macrophage/microglia activation, and nuclear factor (NF)-κB p65 and cyclooxygenase-2 expression in the spinal cord. In vitro, iguratimod treatment inhibited nuclear translocation of NF-κB p65 and down-regulated pro-inflammatory responses in macrophages and microglia. Our results suggest that iguratimod ameliorates acute and chronic EAE by suppressing inflammatory cell infiltration and immune cell activation, partly through inhibition of NF-κB p65, supporting the therapeutic potential of this drug for not only acute, but also chronic MS.
Collapse
Affiliation(s)
- Guangrui Li
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Mei Fang
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hirofumi Ochi
- Department of Geriatric Medicine and Neurology, Ehime University Graduate School of Medicine, Matsuyama, 791-0295, Japan
| | - Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
18
|
Bibert S, Bratschi MW, Aboagye SY, Collinet E, Scherr N, Yeboah-Manu D, Beuret C, Pluschke G, Bochud PY. Susceptibility to Mycobacterium ulcerans Disease (Buruli ulcer) Is Associated with IFNG and iNOS Gene Polymorphisms. Front Microbiol 2017; 8:1903. [PMID: 29046669 PMCID: PMC5632961 DOI: 10.3389/fmicb.2017.01903] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/19/2017] [Indexed: 01/10/2023] Open
Abstract
Buruli ulcer (BU) is a chronic necrotizing disease of the skin and subcutaneous fat tissue. The causative agent, Mycobacterium ulcerans, produces mycolactone, a macrolide toxin, which causes apoptosis of mammalian cells. Only a small proportion of individuals exposed to M. ulcerans develop clinical disease, as surrounding macrophages may control the infection by bacterial killing at an early stage, while mycolactone concentration is still low. Otherwise, bacterial multiplication leads to in higher concentrations of mycolactone, with formation of necrotizing lesions that are no more accessible to immune cells. By typing a cohort of 96 Ghanaian BU patients and 384 endemic controls without BU, we show an association between BU and single nucleotide polymorphisms (SNPs) in iNOS (rs9282799) and IFNG (rs2069705). Both polymorphisms influence promoter activity in vitro. A previously reported SNP in SLC11A1 (NRAMP, rs17235409) tended to be associated with BU. Altogether, these data reflect the importance of IFNG signaling in early defense against M. ulcerans infection.
Collapse
Affiliation(s)
- Stéphanie Bibert
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Martin W Bratschi
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Samuel Y Aboagye
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Emilie Collinet
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Nicole Scherr
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Dorothy Yeboah-Manu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Christian Beuret
- Spiez Laboratory, Federal Office for Civil Protection, Spiez, Switzerland
| | - Gerd Pluschke
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Pierre-Yves Bochud
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Braverman J, Stanley SA. Nitric Oxide Modulates Macrophage Responses to Mycobacterium tuberculosis Infection through Activation of HIF-1α and Repression of NF-κB. THE JOURNAL OF IMMUNOLOGY 2017; 199:1805-1816. [PMID: 28754681 DOI: 10.4049/jimmunol.1700515] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/03/2017] [Indexed: 12/26/2022]
Abstract
IFN-γ is essential for control of Mycobacterium tuberculosis infection in vitro and in vivo. However, the mechanisms by which IFN-γ controls infection remain only partially understood. One of the crucial IFN-γ target genes required for control of M. tuberculosis is inducible NO synthase (iNOS). Although NO produced by iNOS is thought to have direct bactericidal activity against M. tuberculosis, the role of NO as a signaling molecule has been poorly characterized in the context M. tuberculosis infection. In this study, we found that iNOS broadly regulates the macrophage transcriptome during M. tuberculosis infection, activating antimicrobial pathways while also limiting inflammatory cytokine production. The transcription factor hypoxia inducible factor-1α (HIF-1α) was recently shown to be critical for IFN-γ-mediated control of M. tuberculosis infection. We found that HIF-1α function requires NO production, and that HIF-1α and iNOS are linked by a positive feedback loop that amplifies macrophage activation. Furthermore, we found that NO inhibits NF-κB activity to prevent hyperinflammatory responses. Thus, NO activates robust microbicidal programs while also limiting damaging inflammation. IFN-γ signaling must carefully calibrate an effective immune response that does not cause excessive tissue damage, and this study identifies NO as a key player in establishing this balance during M. tuberculosis infection.
Collapse
Affiliation(s)
- Jonathan Braverman
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720; and
| | - Sarah A Stanley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720; and .,Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
20
|
Stortz JA, Raymond SL, Mira JC, Moldawer LL, Mohr AM, Efron PA. Murine Models of Sepsis and Trauma: Can We Bridge the Gap? ILAR J 2017; 58:90-105. [PMID: 28444204 PMCID: PMC5886315 DOI: 10.1093/ilar/ilx007] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 02/20/2017] [Accepted: 02/23/2017] [Indexed: 02/06/2023] Open
Abstract
Sepsis and trauma are both leading causes of death in the United States and represent major public health challenges. Murine models have largely been used in sepsis and trauma research to better understand the pathophysiological changes that occur after an insult and to develop potential life-saving therapeutic agents. Mice are favorable subjects for this type of research given the variety of readily available strains including inbred, outbred, and transgenic strains. In addition, they are relatively easy to maintain and have a high fecundity. However, pharmacological therapies demonstrating promise in preclinical mouse models of sepsis and trauma often fail to demonstrate similar efficacy in human clinical trials, prompting considerable criticism surrounding the capacity of murine models to recapitulate complex human diseases like sepsis and traumatic injury. Fundamental differences between the two species include, but are not limited to, the divergence of the transcriptomic response, the mismatch of temporal response patterns, differences in both innate and adaptive immunity, and heterogeneity within the human population in comparison to the homogeneity of highly inbred mouse strains. Given the ongoing controversy, this narrative review aims to not only highlight the historical importance of the mouse as an animal research model but also highlight the current benefits and limitations of the model as it pertains to sepsis and trauma. Lastly, this review will propose future directions that may promote further use of the model.
Collapse
Affiliation(s)
- Julie A. Stortz
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Steven L. Raymond
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Juan C. Mira
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Lyle L. Moldawer
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Alicia M. Mohr
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Philip A. Efron
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| |
Collapse
|
21
|
Bilham K, Boyd AC, Preston SG, Buesching CD, Newman C, Macdonald DW, Smith AL. Badger macrophages fail to produce nitric oxide, a key anti-mycobacterial effector molecule. Sci Rep 2017; 7:45470. [PMID: 28382943 PMCID: PMC5382539 DOI: 10.1038/srep45470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/01/2017] [Indexed: 12/27/2022] Open
Abstract
The European badger is recognised as a wildlife reservoir for bovine tuberculosis (bTB); the control of which is complex, costly and controversial. Despite the importance of badgers in bTB and the well-documented role for macrophages as anti-mycobacterial effector cells, badger macrophage (bdMφ) responses remain uncharacterised. Here, we demonstrate that bdMφ fail to produce nitric oxide (NO) or upregulate inducible nitric oxide synthase (iNOS) mRNA following Toll-like receptor (TLR) agonist treatment. BdMφ also failed to make NO after stimulation with recombinant badger interferon gamma (bdIFNγ) or a combination of bdIFNγ and lipopolysaccharide. Exposure of bdMφ to TLR agonists and/or bdIFNγ resulted in upregulated cytokine (IL1β, IL6, IL12 and TNFα) mRNA levels indicating that these critical pathways were otherwise intact. Although stimulation with most TLR agonists resulted in strong cytokine mRNA responses, weaker responses were evident after exposure to TLR9 agonists, potentially due to very low expression of TLR9 in bdMφ. Both NO and TLR9 are important elements of innate immunity to mycobacteria, and these features of bdMφ biology would impair their capacity to resist bTB infection. These findings have significant implications for the development of bTB management strategies, and support the use of vaccination to reduce bTB infection in badgers.
Collapse
Affiliation(s)
- Kirstin Bilham
- Department of Zoology, University of Oxford, South Parks Road, OX1 3PS, United Kingdom.,Wildlife Conservation Research Unit Department of Zoology, University of Oxford, The Recanati-Kaplan Centre, Tubney House, Abingdon Road, Tubney, Abingdon OX13 5QL, United Kingdom
| | - Amy C Boyd
- Department of Zoology, University of Oxford, South Parks Road, OX1 3PS, United Kingdom
| | - Stephen G Preston
- Department of Zoology, University of Oxford, South Parks Road, OX1 3PS, United Kingdom
| | - Christina D Buesching
- Wildlife Conservation Research Unit Department of Zoology, University of Oxford, The Recanati-Kaplan Centre, Tubney House, Abingdon Road, Tubney, Abingdon OX13 5QL, United Kingdom
| | - Chris Newman
- Wildlife Conservation Research Unit Department of Zoology, University of Oxford, The Recanati-Kaplan Centre, Tubney House, Abingdon Road, Tubney, Abingdon OX13 5QL, United Kingdom
| | - David W Macdonald
- Wildlife Conservation Research Unit Department of Zoology, University of Oxford, The Recanati-Kaplan Centre, Tubney House, Abingdon Road, Tubney, Abingdon OX13 5QL, United Kingdom
| | - Adrian L Smith
- Department of Zoology, University of Oxford, South Parks Road, OX1 3PS, United Kingdom
| |
Collapse
|
22
|
Lutzke A, Tapia JB, Neufeld MJ, Reynolds MM. Sustained Nitric Oxide Release from a Tertiary S-Nitrosothiol-based Polyphosphazene Coating. ACS APPLIED MATERIALS & INTERFACES 2017; 9:2104-2113. [PMID: 28068065 DOI: 10.1021/acsami.6b12888] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Nitric oxide (NO) occurs naturally in mammalian biochemistry as a critical signaling molecule and exhibits antithrombotic, antibacterial, and wound-healing properties. NO-forming biodegradable polymers have been utilized in the development of antithrombotic or antibacterial materials for biointerfacial applications, including tissue engineering and the fabrication of erodible coatings for medical devices such as stents. Use of such NO-forming polymers has frequently been constrained by short-term release or limited NO storage capacity and has led to the pursuit of new materials with improved NO release function. Herein, we report the development of an NO-releasing bioerodible coating prepared from poly[bis(3-mercapto-3-methylbut-1-yl glycinyl)phosphazene] (POP-Gly-MMB), a polyphosphazene based on glycine and the naturally occurring tertiary thiol 3-mercapto-3-methylbutan-1-ol (MMB). To evaluate the NO release properties of this material, the thiolated polymer POP-Gly-MMB-SH was applied as a coating to glass substrates and subsequently converted to the NO-forming S-nitrosothiol (RSNO) derivative (POP-Gly-MMB-NO) by immersion in a mixture of tert-butyl nitrite (t-BuONO) and pentane. NO release flux from the coated substrates was determined by chemiluminescence-based NO measurement and was found to remain in a physiologically relevant range for up to 2 weeks (6.5-0.090 nmol of NO·min-1·cm-2) when immersed in pH 7.4 phosphate-buffered saline (PBS) at 37 °C. Furthermore, the coating exhibited an overall NO storage capacity of 0.89 ± 0.09 mmol·g-1 (4.3 ± 0.6 μmol·cm-2). Erosion of POP-Gly-MMB-NO in PBS at 37 °C over 6 weeks results in 14% mass loss, and time-of-flight mass spectrometry (TOF-MS) was used to characterize the organic products of hydrolytic degradation as glycine, MMB, and several related esters. The comparatively long-term NO release and high storage capacity of POP-Gly-MMB-NO coatings suggest potential as a source of therapeutic NO for biomedical applications.
Collapse
Affiliation(s)
- Alec Lutzke
- Department of Chemistry, ‡School of Biomedical Engineering, and §Department of Chemical & Biological Engineering, Colorado State University , Fort Collins, Colorado 80523, United States
| | - Jesus B Tapia
- Department of Chemistry, ‡School of Biomedical Engineering, and §Department of Chemical & Biological Engineering, Colorado State University , Fort Collins, Colorado 80523, United States
| | - Megan J Neufeld
- Department of Chemistry, ‡School of Biomedical Engineering, and §Department of Chemical & Biological Engineering, Colorado State University , Fort Collins, Colorado 80523, United States
| | - Melissa M Reynolds
- Department of Chemistry, ‡School of Biomedical Engineering, and §Department of Chemical & Biological Engineering, Colorado State University , Fort Collins, Colorado 80523, United States
| |
Collapse
|
23
|
Cardiovascular risk assessment in patients with rheumatoid arthritis: The relevance of clinical, genetic and serological markers. Autoimmun Rev 2016; 15:1013-1030. [DOI: 10.1016/j.autrev.2016.07.026] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/09/2016] [Indexed: 12/11/2022]
|
24
|
Fattakhov NS, Vasilenko MA, Skuratovskaia DA, Kulikov DI, Kirienkova EV, Zatolokin PA, Beletskaya MA, Litvinova LS. [Pathogenetic significance of C774T single nucleotide polymorphism of the endothelial NO synthase gene in the development of metabolic syndrome]. BIOMEDITSINSKAIA KHIMIIA 2016; 62:447-52. [PMID: 27562999 DOI: 10.18097/pbmc20166204447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The relationship between nitric oxide production and metabolic disorders and the role of endothelial nitric oxide synthase (eNOS or NOS3) in metabolic syndrome (MS) remain poorly understood and need deeper investigation. In this context the role of the NOS3 gene in pathogenesis of MS is of special interest. The aim of the study was to investigate association of NOS3 single nucleotide polymorphism C774T with risk of MS in the Slavic population of the Kaliningrad region and the relationship of this polymorphic variant with some parameters of endothelial dysfunction. The study included 128 patients (48 men and 80 women aged from 36 to 52 years) with MS. The control group consisted of 126 healthy volunteers (60 men and 66 women aged from 30 to 40 years). Genotyping was performed by real-time PCR. Serum nitrite levels were determined spectrophotometrically by the Griess method. Serum levels of endothelin-1 and eNOS were evaluated by ELISA. The study has shown association of T allele (OR=2.06; p=0.0004; CI: 1.38-3.08) and CT genotype (OR=1.97; p=0.014; CI: 1.14-3.40 ) C774T polymorphism of the NOS3 gene with risk of MS in the Slavic population of the Kaliningrad region. Allele C (OR=0.48; p=0.0004; CI: 0.32-0.72) and homozygous CC genotype (OR=0.41; p=0.001; CI: 0.24-0,69) C774T polymorphism of the NOS3 gene were associated with reduced risk of the development of MS. Significant differences in serum levels of eNOS and endothelin-1 depended on the CT and TT genotypes of C774T polymorphism of the NOS3 gene in MS.
Collapse
Affiliation(s)
- N S Fattakhov
- Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - M A Vasilenko
- Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | | | - D I Kulikov
- Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - E V Kirienkova
- Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - P A Zatolokin
- Regional Clinical Hospital of the Kaliningrad Region, Kaliningrad, Russia
| | - M A Beletskaya
- Municipal Children's Hospital No. 6, Kaliningrad, Russia
| | - L S Litvinova
- Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| |
Collapse
|
25
|
Tallam A, Perumal TM, Antony PM, Jäger C, Fritz JV, Vallar L, Balling R, del Sol A, Michelucci A. Gene Regulatory Network Inference of Immunoresponsive Gene 1 (IRG1) Identifies Interferon Regulatory Factor 1 (IRF1) as Its Transcriptional Regulator in Mammalian Macrophages. PLoS One 2016; 11:e0149050. [PMID: 26872335 PMCID: PMC4752512 DOI: 10.1371/journal.pone.0149050] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/25/2016] [Indexed: 01/28/2023] Open
Abstract
Immunoresponsive gene 1 (IRG1) is one of the highest induced genes in macrophages under pro-inflammatory conditions. Its function has been recently described: it codes for immune-responsive gene 1 protein/cis-aconitic acid decarboxylase (IRG1/CAD), an enzyme catalysing the production of itaconic acid from cis-aconitic acid, a tricarboxylic acid (TCA) cycle intermediate. Itaconic acid possesses specific antimicrobial properties inhibiting isocitrate lyase, the first enzyme of the glyoxylate shunt, an anaplerotic pathway that bypasses the TCA cycle and enables bacteria to survive on limited carbon conditions. To elucidate the mechanisms underlying itaconic acid production through IRG1 induction in macrophages, we examined the transcriptional regulation of IRG1. To this end, we studied IRG1 expression in human immune cells under different inflammatory stimuli, such as TNFα and IFNγ, in addition to lipopolysaccharides. Under these conditions, as previously shown in mouse macrophages, IRG1/CAD accumulates in mitochondria. Furthermore, using literature information and transcription factor prediction models, we re-constructed raw gene regulatory networks (GRNs) for IRG1 in mouse and human macrophages. We further implemented a contextualization algorithm that relies on genome-wide gene expression data to infer putative cell type-specific gene regulatory interactions in mouse and human macrophages, which allowed us to predict potential transcriptional regulators of IRG1. Among the computationally identified regulators, siRNA-mediated gene silencing of interferon regulatory factor 1 (IRF1) in macrophages significantly decreased the expression of IRG1/CAD at the gene and protein level, which correlated with a reduced production of itaconic acid. Using a synergistic approach of both computational and experimental methods, we here shed more light on the transcriptional machinery of IRG1 expression and could pave the way to therapeutic approaches targeting itaconic acid levels.
Collapse
Affiliation(s)
- Aravind Tallam
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Thaneer M. Perumal
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Paul M. Antony
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Christian Jäger
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Joëlle V. Fritz
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Laurent Vallar
- Genomics Research Laboratory, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Antonio del Sol
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alessandro Michelucci
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
26
|
Grishin A, Bowling J, Bell B, Wang J, Ford HR. Roles of nitric oxide and intestinal microbiota in the pathogenesis of necrotizing enterocolitis. J Pediatr Surg 2016; 51:13-7. [PMID: 26577908 PMCID: PMC4894644 DOI: 10.1016/j.jpedsurg.2015.10.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/06/2015] [Indexed: 12/12/2022]
Abstract
Necrotizing enterocolitis remains one of the most vexing problems in the neonatal intensive care unit. Risk factors for NEC include prematurity, formula feeding, and inappropriate microbial colonization of the GI tract. The pathogenesis of NEC is believed to involve weakening of the intestinal barrier by perinatal insults, translocation of luminal bacteria across the weakened barrier, an exuberant inflammatory response, and exacerbation of the barrier damage by inflammatory factors, leading to a vicious cycle of inflammation-inflicted epithelial damage. Nitric oxide (NO), produced by inducible NO synthase (iNOS) and reactive NO oxidation intermediates play a prominent role in the intestinal barrier damage by inducing enterocyte apoptosis and inhibiting the epithelial restitution processes, namely enterocyte proliferation and migration. The factors that govern iNOS upregulation in the intestine are not well understood, which hampers efforts in developing NO/iNOS-targeted therapies. Similarly, efforts to identify bacteria or bacterial colonization patterns associated with NEC have met with limited success, because the same bacterial species can be found in NEC and in non-NEC subjects. However, microbiome studies have identified the three important characteristics of early bacterial populations of the GI tract: high diversity, low complexity, and fluidity. Whether NEC is caused by specific bacteria remains a matter of debate, but data from hospital outbreaks of NEC strongly argue in favor of the infectious nature of this disease. Studies in Cronobacter muytjensii have established that the ability to induce NEC is the property of specific strains rather than the species as a whole. Progress in our understanding of the roles of bacteria in NEC will require microbiological experiments and genome-wide analysis of virulence factors.
Collapse
Affiliation(s)
- Anatoly Grishin
- Division of Pediatric Surgery, Children's Hospital Los Angeles, 4650 Sunset Boulevard, Los Angeles, CA 90027; Department of Surgery, Keck School of Medicine of the University of Southern California, 4650 Sunset Boulevard, Los Angeles, CA 90027.
| | - Jordan Bowling
- Department of Surgery, Keck School of Medicine of the University of Southern California, 4650 Sunset Boulevard, Los Angeles, CA 90027.
| | - Brandon Bell
- Division of Pediatric Surgery, Children's Hospital Los Angeles, 4650 Sunset Boulevard, Los Angeles, CA 90027.
| | - Jin Wang
- Division of Pediatric Surgery, Children's Hospital Los Angeles, 4650 Sunset Boulevard, Los Angeles, CA 90027.
| | - Henri R Ford
- Division of Pediatric Surgery, Children's Hospital Los Angeles, 4650 Sunset Boulevard, Los Angeles, CA 90027; Department of Surgery, Keck School of Medicine of the University of Southern California, 4650 Sunset Boulevard, Los Angeles, CA 90027.
| |
Collapse
|
27
|
Abstract
Apart from its classical function in bone and calcium metabolism, vitamin D is also involved in immune regulation and has been linked to various cancers, immune disorders and allergic diseases. Within the innate and adaptive immune systems, the vitamin D receptor and enzymes in monocytes, dendritic cells, epithelial cells, T lymphocytes and B lymphocytes mediate the immune modulatory actions of vitamin D. Vitamin D insufficiency/deficiency early in life has been identified as one of the risk factors for food allergy. Several studies have observed an association between increasing latitude and food allergy prevalence, plausibly linked to lower ultraviolet radiation (UVR) exposure and vitamin D synthesis in the skin. Along with mounting epidemiological evidence of a link between vitamin D status and food allergy, mice and human studies have shed light on the modulatory properties of vitamin D on the innate and adaptive immune systems. This review will summarize the literature on the metabolism and immune modulatory properties of vitamin D, with particular reference to food allergy.
Collapse
|
28
|
Cho YC, Ju A, Kim BR, Cho S. Anti-inflammatory effects of Crataeva nurvala Buch. Ham. are mediated via inactivation of ERK but not NF-κB. JOURNAL OF ETHNOPHARMACOLOGY 2015; 162:140-147. [PMID: 25571844 DOI: 10.1016/j.jep.2014.12.056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 12/02/2014] [Accepted: 12/24/2014] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Crataeva nurvala Buch. Ham. is an important medicinal plant in India, and its extracts and components were used to treat various inflammatory diseases, such as urinary tract infection, rheumatoid arthritis, and colitis. However, no systemic studies about anti-inflammatory effects of Crataeva nurvala Buch. Ham. and its underlying mechanisms of action have been reported. This study aimed to explore the anti-inflammatory effects of ethanol extracts of Crataeva nurvala Buch. Ham. (ECN). MATERIALS AND METHODS The non-cytotoxic and maximal effective concentration of ECN was determined by measuring the formation of formazan from water-soluble tetrazolium salt in living cells. The inhibitory effect of ECN on nitric oxide (NO) synthesis was measured using Griess reagent, and Enzyme-linked immunosorbent assay (ELISA) was used to measure secreted tumor necrosis factor (TNF)-α and interleukin (IL)-6 protein levels. Furthermore, reverse transcription polymerase chain reaction (RT-PCR) and Western blotting analysis were used to assess the mRNA and protein expression of each inflammatory mediator or relating signaling protein, respectively. RESULTS A non-cytotoxic concentration of ECN (≤200 μg/ml) significantly reduced the production of NO and IL-6, but not TNF-α, in lipopolysaccharides (LPS)-stimulated RAW 264.7 macrophages. Decreased production of NO by ECN was correlated with reduced expression of iNOS at the mRNA and protein levels. However, cyclooxygenase (COX)-2 expressions at mRNA and protein level were not regulated by ECN. The mRNA expression of IL-6 and IL-1β, but not TNF-α, was also inhibited by ECN treatment in LPS-stimulated RAW 264.7 macrophages. Reduced production of inflammatory mediators by ECN was followed by decreased activity of mitogen-activated protein kinases (MAPKs), especially extracellular signal-regulated kinase (ERK), but not nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). CONCLUSIONS These results indicate that ECN inhibits LPS-induced inflammatory responses via negative regulation of ERK in murine macrophages, suggesting that ECN is a candidate for alleviating severe inflammation.
Collapse
Affiliation(s)
- Young-Chang Cho
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| | - Anna Ju
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| | - Ba Reum Kim
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| | - Sayeon Cho
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea.
| |
Collapse
|
29
|
Rajaram MVS, Ni B, Dodd CE, Schlesinger LS. Macrophage immunoregulatory pathways in tuberculosis. Semin Immunol 2014; 26:471-85. [PMID: 25453226 DOI: 10.1016/j.smim.2014.09.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/17/2022]
Abstract
Macrophages, the major host cells harboring Mycobacterium tuberculosis (M.tb), are a heterogeneous cell type depending on their tissue of origin and host they are derived from. Significant discord in macrophage responses to M.tb exists due to differences in M.tb strains and the various types of macrophages used to study tuberculosis (TB). This review will summarize current concepts regarding macrophage responses to M.tb infection, while pointing out relevant differences in experimental outcomes due to the use of divergent model systems. A brief description of the lung environment is included since there is increasing evidence that the alveolar macrophage (AM) has immunoregulatory properties that can delay optimal protective host immune responses. In this context, this review focuses on selected macrophage immunoregulatory pattern recognition receptors (PRRs), cytokines, negative regulators of inflammation, lipid mediators and microRNAs (miRNAs).
Collapse
Affiliation(s)
- Murugesan V S Rajaram
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Bin Ni
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Claire E Dodd
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
30
|
Mattila JT, Thomas AC. Nitric oxide synthase: non-canonical expression patterns. Front Immunol 2014; 5:478. [PMID: 25346730 PMCID: PMC4191211 DOI: 10.3389/fimmu.2014.00478] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/19/2014] [Indexed: 12/12/2022] Open
Abstract
Science can move ahead by questioning established or canonical views and, so it may be with the enzymes, nitric oxide synthases (NOS). Nitric oxide (NO) is generated by NOS isoforms that are often described by their tissue-specific expression patterns. NOS1 (nNOS) is abundant in neural tissue, NOS2 is upregulated in activated macrophages and known as inducible NOS (iNOS), and NOS3 (eNOS) is abundant in endothelium where it regulates vascular tone. These isoforms are described as constitutive or inducible, but in this perspective we question the broad application of these labels. Are there instances where "constitutive" NOS (NOS1 and NOS3) are inducibly expressed; conversely, are there instances where NOS2 is constitutively expressed? NOS1 and NOS3 inducibility may be linked to post-translational regulation, making their actual patterns activity much more difficult to detect. Constitutive NOS2 expression has been observed in several tissues, especially the human pulmonary epithelium where it may regulate airway tone. These data suggest that expression of the three NOS enzymes may include non-established patterns. Such information should be useful in designing strategies to modulate these important enzymes in different disease states.
Collapse
Affiliation(s)
- Joshua T. Mattila
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anita C. Thomas
- Bristol Heart Institute and Bristol CardioVascular, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| |
Collapse
|
31
|
Thomas AC, Mattila JT. "Of mice and men": arginine metabolism in macrophages. Front Immunol 2014; 5:479. [PMID: 25339954 PMCID: PMC4188127 DOI: 10.3389/fimmu.2014.00479] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/19/2014] [Indexed: 01/07/2023] Open
Affiliation(s)
- Anita C Thomas
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol , Bristol , UK
| | - Joshua T Mattila
- Department of Microbiology and Molecular Genetics, University of Pittsburgh , Pittsburgh, PA , USA
| |
Collapse
|
32
|
Study of the correlations among some parameters of the oxidative status, gelatinases, and their inhibitors in a group of subjects with metabolic syndrome. Mediators Inflamm 2014; 2014:510619. [PMID: 25114377 PMCID: PMC4121250 DOI: 10.1155/2014/510619] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/26/2014] [Accepted: 06/27/2014] [Indexed: 12/14/2022] Open
Abstract
Our aim was to examine some parameters of oxidative status, gelatinases, and their inhibitors and to evaluate their interrelationships in subjects with metabolic syndrome (MS). We enrolled 65 MS subjects, subdivided according to the presence or not of diabetes mellitus. We examined lipid peroxidation (expressed as thiobarbituric acid reacting substances, TBARS), protein oxidation (expressed as carbonyl groups), nitric oxide metabolites (NOx), total antioxidant status (TAS), MMP-2, MMP-9, TIMP-1, and TIMP-2. We found that MS subjects, diabetics and nondiabetics, showed an increase in TBARS, PC, and NOx. A significant decrease in TAS was observed only in nondiabetic MS subjects in comparison with diabetic MS subjects. We observed increased concentrations of MMP-2, MMP-9, TIMP-1, and TIMP-2, higher in diabetic subjects. Our data showed a positive correlation between TAS and MMP-2, TAS and MMP-9, and TAS and MMP-9/TIMP-1 and a negative correlation between TBARS and MMP-2 in diabetic MS subjects in the entire group. In MS subjects a prooxidant status and increased levels of gelatinases and their inhibitors are evident although the correlations between oxidative stress and MMPs or TIMPs are controversial and need further investigation.
Collapse
|
33
|
Lipid peroxidation, nitric oxide metabolites, and their ratio in a group of subjects with metabolic syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:824756. [PMID: 24987495 PMCID: PMC4060162 DOI: 10.1155/2014/824756] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/13/2014] [Accepted: 05/17/2014] [Indexed: 01/11/2023]
Abstract
Our aim was to evaluate lipid peroxidation, expressed as thiobarbituric acid-reactive substances (TBARS), nitric oxide metabolites (nitrite + nitrate) expressed as NOx, and TBARS/NOx ratio in a group of subjects with metabolic syndrome (MS). In this regard we enrolled 106 subjects with MS defined according to the IDF criteria, subsequently subdivided into diabetic (DMS) and nondiabetic (NDMS) and also into subjects with a low triglycerides/HDL-cholesterol (TG/HDL-C) index or with a high TG/HDL-C index. In the entire group and in the four subgroups of MS subjects we found an increase in TBARS and NOx levels and a decrease in TBARS/NOx ratio in comparison with normal controls. Regarding all these parameters no statistical difference between DMS and NDMS was evident, but a significant increase in NOx was present in subjects with a high TG/HDL-C index in comparison with those with a low index. In MS subjects we also found a negative correlation between TBARS/NO x ratio and TG/HDL-C index. Considering the hyperactivity of the inducible NO synthase in MS, these data confirm the altered redox and inflammatory status that characterizes the MS and suggest a link between lipid peroxidation, inflammation, and insulin resistance, evaluated as TG/HDL-C index.
Collapse
|
34
|
Gross TJ, Kremens K, Powers LS, Brink B, Knutson T, Domann FE, Philibert RA, Milhem MM, Monick MM. Epigenetic silencing of the human NOS2 gene: rethinking the role of nitric oxide in human macrophage inflammatory responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:2326-38. [PMID: 24477906 PMCID: PMC3943971 DOI: 10.4049/jimmunol.1301758] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Macrophages, including alveolar macrophages, are primary phagocytic cells of the innate immune system. Many studies of macrophages and inflammation have been done in mouse models, in which inducible NO synthase (NOS2) and NO are important components of the inflammatory response. Human macrophages, in contrast to mouse macrophages, express little detectable NOS2 and generate little NO in response to potent inflammatory stimuli. The human NOS2 gene is highly methylated around the NOS2 transcription start site. In contrast, mouse macrophages contain unmethylated cytosine-phosphate-guanine (CpG) dinucleotides proximal to the NOS2 transcription start site. Further analysis of chromatin accessibility and histone modifications demonstrated a closed conformation at the human NOS2 locus and an open conformation at the murine NOS2 locus. In examining the potential for CpG demethylation at the NOS2 locus, we found that the human NOS2 gene was resistant to the effects of demethylation agents both in vitro and in vivo. Our data demonstrate that epigenetic modifications in human macrophages are associated with CpG methylation, chromatin compaction, and histone modifications that effectively silence the NOS2 gene. Taken together, our findings suggest there are significant and underappreciated differences in how murine and human macrophages respond to inflammatory stimuli.
Collapse
Affiliation(s)
- Thomas J. Gross
- Department of Medicine, Carver College of Medicine, The
University of Iowa, Iowa City, Iowa, 52242
| | - Karol Kremens
- Department of Medicine, Carver College of Medicine, The
University of Iowa, Iowa City, Iowa, 52242
| | - Linda S. Powers
- Department of Medicine, Carver College of Medicine, The
University of Iowa, Iowa City, Iowa, 52242
| | - Brandi Brink
- Department of Medicine, Carver College of Medicine, The
University of Iowa, Iowa City, Iowa, 52242
| | - Tina Knutson
- Department of Medicine, Carver College of Medicine, The
University of Iowa, Iowa City, Iowa, 52242
| | - Frederick E. Domann
- Department of Radiation Oncology, Carver College of
Medicine, The University of Iowa, Iowa City, Iowa, 52242
| | - Robert A. Philibert
- Department of Psychiatry, Carver College of Medicine, The
University of Iowa, Iowa City, Iowa, 52242
| | - Mohammed M. Milhem
- Department of Medicine, Carver College of Medicine, The
University of Iowa, Iowa City, Iowa, 52242
| | - Martha M. Monick
- Department of Medicine, Carver College of Medicine, The
University of Iowa, Iowa City, Iowa, 52242
| |
Collapse
|
35
|
Effects of dietary nutrients on volatile breath metabolites. J Nutr Sci 2013; 2:e34. [PMID: 25191584 PMCID: PMC4153095 DOI: 10.1017/jns.2013.26] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 07/22/2013] [Accepted: 07/25/2013] [Indexed: 12/22/2022] Open
Abstract
Breath analysis is becoming increasingly established as a means of assessing metabolic,
biochemical and physiological function in health and disease. The methods available for
these analyses exploit a variety of complex physicochemical principles, but are becoming
more easily utilised in the clinical setting. Whilst some of the factors accounting for
the biological variation in breath metabolite concentrations have been clarified, there
has been relatively little work on the dietary factors that may influence them. In
applying breath analysis to the clinical setting, it will be important to consider how
these factors may affect the interpretation of endogenous breath composition. Diet may
have complex effects on the generation of breath compounds. These effects may either be
due to a direct impact on metabolism, or because they alter the gastrointestinal flora.
Bacteria are a major source of compounds in breath, and their generation of H2,
hydrogen cyanide, aldehydes and alkanes may be an indicator of the health of their
host.
Collapse
|
36
|
Brüne B, Dehne N, Grossmann N, Jung M, Namgaladze D, Schmid T, von Knethen A, Weigert A. Redox control of inflammation in macrophages. Antioxid Redox Signal 2013; 19:595-637. [PMID: 23311665 PMCID: PMC3718318 DOI: 10.1089/ars.2012.4785] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 12/14/2012] [Accepted: 01/11/2013] [Indexed: 12/13/2022]
Abstract
Macrophages are present throughout the human body, constitute important immune effector cells, and have variable roles in a great number of pathological, but also physiological, settings. It is apparent that macrophages need to adjust their activation profile toward a steadily changing environment that requires altering their phenotype, a process known as macrophage polarization. Formation of reactive oxygen species (ROS), derived from NADPH-oxidases, mitochondria, or NO-producing enzymes, are not necessarily toxic, but rather compose a network signaling system, known as redox regulation. Formation of redox signals in classically versus alternatively activated macrophages, their action and interaction at the level of key targets, and the resulting physiology still are insufficiently understood. We review the identity, source, and biological activities of ROS produced during macrophage activation, and discuss how they shape the key transcriptional responses evoked by hypoxia-inducible transcription factors, nuclear-erythroid 2-p45-related factor 2 (Nrf2), and peroxisome proliferator-activated receptor-γ. We summarize the mechanisms how redox signals add to the process of macrophage polarization and reprogramming, how this is controlled by the interaction of macrophages with their environment, and addresses the outcome of the polarization process in health and disease. Future studies need to tackle the option whether we can use the knowledge of redox biology in macrophages to shape their mediator profile in pathophysiology, to accelerate healing in injured tissue, to fight the invading pathogens, or to eliminate settings of altered self in tumors.
Collapse
Affiliation(s)
- Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I-Pathobiochemistry, Goethe-University Frankfurt, Frankfurt, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Interactions between an M. tuberculosis strain overexpressing mtrA and mononuclear phagocytes. Adv Med Sci 2013; 58:172-83. [PMID: 23640943 DOI: 10.2478/v10039-012-0058-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE It was previously shown that the bacterial two-component regulatory signal transduction (2CR) system MtrAB may be associated with the ability of M. tuberculosis (Mtb) to survive in macrophages. In the present work Mtb mutants: Rv-78 with overexpression of mtrA and Rv-129 with elevated level of phosphorylation-defective MtrA were used for further investigation of the potential influence of the MtrAB system on Mtb interaction with human monocytes. MATERIAL/METHODS Flow cytometry was used to determine the expression of MHC class II molecules. The expression of genes for inducible nitric oxide synthase (iNOS) and cathepsin G was quantified by RT-PCR. The association of Mtb strains with Rab5 and Rab7 positive vacuoles was investigated applying confocal microscopy. IL-10 and IL-12 secretion by monocytes as well as the Mtb susceptibility to cathepsin G were investigated. RESULTS Mutation-carried and wild type Mtb strains inhibited MHC class II expression on monocytes to a similar extent. Monocyte stimulation with mycobacteria led to the increased production of IL-10 but no detectable amounts of IL-12 or NO were observed. Expression of the gene for iNOS was not detected while that for cathepsin G was shown, however its intensity was not associated with MtrA mutation. Mtb mutant strains were more effectively enclosed in phagosomes containing the late endosome marker Rab7 as compared to the control. CONCLUSIONS The results may confirm the importance of the MtrAB system in mycobacterial capacity for successful survival in phagocytes, especially in the context of high degree of colocalization of Mtb Rv-78 to mature phagosomes.
Collapse
|
38
|
Carneiro LA, Laurenti MD, Campos MB, Gomes CMDC, Corbett CEP, Silveira FT. Susceptibility of peritoneal macrophage from different species of neotropical primates to ex vivo Leishmania (L.) infantum chagasi-infection. Rev Inst Med Trop Sao Paulo 2012; 54:95-101. [PMID: 22499423 DOI: 10.1590/s0036-46652012000200007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Indexed: 11/22/2022] Open
Abstract
This study examined the susceptibility of peritoneal macrophage (PM) from the Neotropical primates: Callithrix jacchus, Callithrix penicillata, Saimiri sciureus, Aotus azarae infulatus and Callimico goeldii to ex vivo Leishmania (L.) infantum chagasi-infection, the etiological agent of American visceral leishmaniasis (AVL), as a screening assay for evaluating the potential of these non-human primates as experimental models for studying AVL. The PM-susceptibility to infection was accessed by the PM-infection index (PMI) at 24, 72 h and by the mean of these rates (FPMI), as well as by the TNF-α, IL-12 (Capture ELISA) and Nitric oxide (NO) responses (Griess method). At 24h, the PMI of A. azarae infulatus (128) was higher than those of C. penicillata (83), C. goeldii (78), S. sciureus (77) and C. jacchus (55). At 72h, there was a significant PMI decrease in four monkeys: A. azarae infulatus (128/37), C. penicillata (83/38), S. sciureus (77/38) and C. jacchus (55/12), with exception of C. goeldii (78/54). The FPMI of A. azarae infulatus (82.5) and C. goeldii (66) were higher than C. jacchus (33.5), but not higher than those of C. penicillata (60.5) and S. sciureus (57.5). The TNF-a response was more regular in those four primates which decreased their PMI at 24/72 h: C. jacchus (145/122 pg/mL), C. penicillata (154/130 pg/mL), S. sciureus (164/104 pg/mL) and A. azarae infulatus (154/104 pg/mL), with exception of C. goeldii (38/83 pg/mL). The IL-12 response was mainly prominent in A. infulatus and C. goeldii which presented the highest FPMI and, the NO response was higher in C. goeldii, mainly at 72 h. These findings strongly suggest that these New World primates have developed a resistant innate immune response mechanism capable of controlling the macrophage intracellular growth of L. (L.) i. chagasi-infection, which do not encourage their use as animal model for studying AVL.
Collapse
Affiliation(s)
- Liliane Almeida Carneiro
- Evandro Chagas Institute, Surveillance Secretary of Health, Ministry of Health, Belém, Pará State, Brazil.
| | | | | | | | | | | |
Collapse
|
39
|
Caimi G, Hopps E, Montana M, Noto D, Canino B, Lo Presti R, Averna MR. Evaluation of nitric oxide metabolites in a group of subjects with metabolic syndrome. Diabetes Metab Syndr 2012; 6:132-135. [PMID: 23158975 DOI: 10.1016/j.dsx.2012.09.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
AIM To evaluate the concentration of metabolites (NO(2)(-), NO(3)(-)) of nitric oxide (NO) in metabolic syndrome (MS). MATERIALS AND METHODS We enrolled 106 subjects (45 women and 61 men) with MS of which 43 (14 women and 27 men) with diabetes mellitus and 63 (31 women and 32 men) without diabetes mellitus, and 54 subjects (19 women and 35 men) as control group. The nitric oxide metabolites (nitrite+nitrate=NOx) were evaluated employing the Griess reagent. RESULTS In the whole group of MS subjects was evident, in comparison with control group, a significant increase in NOx. The same finding was also present between control group and diabetic subjects with MS and between control group and nondiabetic subjects with MS. No difference was observed between the two subgroups (diabetic and nondiabetic subjects with MS) about NOx. Contrasting information were obtained examining the linear regression among NOx, age, anthropometric profile, blood pressure values and glycometabolic pattern of subjects with MS. CONCLUSIONS In MS subjects we found a significant increase in NOx not influenced by diabetes mellitus. The NOx is a parameter that must be considered in MS keeping in mind that its behavior is related to chronic inflammation that accompanies this clinical condition.
Collapse
Affiliation(s)
- Gregorio Caimi
- Dipartimento di Medicina Interna e Specialistica, Università di Palermo, Italy
| | | | | | | | | | | | | |
Collapse
|
40
|
Qidwai T, Jamal F. Inducible nitric oxide synthase (iNOS) gene polymorphism and disease prevalence. Scand J Immunol 2011; 72:375-87. [PMID: 21039732 DOI: 10.1111/j.1365-3083.2010.02458.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nitric oxide synthase gene is present on chromosome 17 and has been implicated in a wide variety of diseases. The nitric oxide synthase enzyme forms nitric oxide that besides being a signalling molecule plays an important role in host immune response. Inducible nitric oxide synthase expression is regulated at the level of transcription. Single-nucleotide polymorphisms, copy number variation and simple sequence repeat are important variations that have been reported in human genome. The presence of such variations in the regulatory region affects the level of gene product in the cell, while variation in the coding region influences the structure of proteins and its activity. This alteration in the level of gene product and the structure of the protein molecule might be responsible for the final outcome of genetic as well as infectious diseases. In the present manuscript, we review the role of inducible nitric oxide synthase (iNOS) gene polymorphisms in different diseases and populations. The iNOS gene with one pentanucleotide repeat, two single-nucleotide polymorphisms in promoter region and one polymorphism in exon 16 has been implicated in several diseases. We have also predicted several polymorphisms in the promoter region of iNOS computationally, which might affect the transcription factor binding site (TFBS) and hypothesize that these polymorphisms have some putative role in the outcome of disease(s).
Collapse
Affiliation(s)
- T Qidwai
- Department of Biochemistry, Dr. R.M.L. Avadh University, Faizabad, India
| | | |
Collapse
|
41
|
Wink DA, Hines HB, Cheng RYS, Switzer CH, Flores-Santana W, Vitek MP, Ridnour LA, Colton CA. Nitric oxide and redox mechanisms in the immune response. J Leukoc Biol 2011; 89:873-91. [PMID: 21233414 DOI: 10.1189/jlb.1010550] [Citation(s) in RCA: 499] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The role of redox molecules, such as NO and ROS, as key mediators of immunity has recently garnered renewed interest and appreciation. To regulate immune responses, these species trigger the eradication of pathogens on the one hand and modulate immunosuppression during tissue-restoration and wound-healing processes on the other. In the acidic environment of the phagosome, a variety of RNS and ROS is produced, thereby providing a cauldron of redox chemistry, which is the first line in fighting infection. Interestingly, fluctuations in the levels of these same reactive intermediates orchestrate other phases of the immune response. NO activates specific signal transduction pathways in tumor cells, endothelial cells, and monocytes in a concentration-dependent manner. As ROS can react directly with NO-forming RNS, NO bioavailability and therefore, NO response(s) are changed. The NO/ROS balance is also important during Th1 to Th2 transition. In this review, we discuss the chemistry of NO and ROS in the context of antipathogen activity and immune regulation and also discuss similarities and differences between murine and human production of these intermediates.
Collapse
Affiliation(s)
- David A Wink
- Radiation Biology Branch, National Cancer Institute/National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Human myeloperoxidase in innate and acquired immunity. Arch Biochem Biophys 2010; 500:92-106. [DOI: 10.1016/j.abb.2010.04.008] [Citation(s) in RCA: 187] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 04/07/2010] [Accepted: 04/12/2010] [Indexed: 12/12/2022]
|
43
|
Ireland R, Olivares-Zavaleta N, Warawa JM, Gherardini FC, Jarrett C, Hinnebusch BJ, Belisle JT, Fairman J, Bosio CM. Effective, broad spectrum control of virulent bacterial infections using cationic DNA liposome complexes combined with bacterial antigens. PLoS Pathog 2010; 6:e1000921. [PMID: 20523903 PMCID: PMC2877747 DOI: 10.1371/journal.ppat.1000921] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 04/23/2010] [Indexed: 12/26/2022] Open
Abstract
Protection against virulent pathogens that cause acute, fatal disease is often hampered by development of microbial resistance to traditional chemotherapeutics. Further, most successful pathogens possess an array of immune evasion strategies to avoid detection and elimination by the host. Development of novel, immunomodulatory prophylaxes that target the host immune system, rather than the invading microbe, could serve as effective alternatives to traditional chemotherapies. Here we describe the development and mechanism of a novel pan-anti-bacterial prophylaxis. Using cationic liposome non-coding DNA complexes (CLDC) mixed with crude F. tularensis membrane protein fractions (MPF), we demonstrate control of virulent F. tularensis infection in vitro and in vivo. CLDC+MPF inhibited bacterial replication in primary human and murine macrophages in vitro. Control of infection in macrophages was mediated by both reactive nitrogen species (RNS) and reactive oxygen species (ROS) in mouse cells, and ROS in human cells. Importantly, mice treated with CLDC+MPF 3 days prior to challenge survived lethal intranasal infection with virulent F. tularensis. Similarly to in vitro observations, in vivo protection was dependent on the presence of RNS and ROS. Lastly, CLDC+MPF was also effective at controlling infections with Yersinia pestis, Burkholderia pseudomallei and Brucella abortus. Thus, CLDC+MPF represents a novel prophylaxis to protect against multiple, highly virulent pathogens. Conventional treatment of bacterial infections typically includes administration of antibiotics. However, many pathogens have developed spontaneous resistance to commonly used antibiotics. Development of new compounds that stimulate the host immune system to directly kill bacteria by mechanisms different from those utilized by antibiotics may serve as effective alternatives to antibiotic therapy. In this report, we describe a novel compound capable of controlling infections mediated by different, unrelated bacteria via the induction of host derived reactive oxygen and reactive nitrogen species. This compound is comprised of cationic liposome DNA complexes (CLDC) and crude membrane preparations (MPF) obtained from attenuated Francisella tularensis Live Vaccine Strain (LVS). Pretreatment of primary mouse or human cells limited replication of virulent F. tularensis, Burkholderia pseudomallei, Yersinia pestis and Brucella abortus in vitro. CLDC+MPF was also effective for controlling lethal pulmonary infections with virulent F. tularensis. Thus, CLDC+MPF represents a novel antimicrobial for treatment of lethal, acute, bacterial infections.
Collapse
Affiliation(s)
- Robin Ireland
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Norma Olivares-Zavaleta
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Jonathan M. Warawa
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Frank C. Gherardini
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Clayton Jarrett
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - B. Joseph Hinnebusch
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - John T. Belisle
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jeffery Fairman
- Juvaris Biotherapeutics, Burlingame, California, United States of America
| | - Catharine M. Bosio
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
- * E-mail:
| |
Collapse
|
44
|
Moheno P, Morrey J, Fuchs D. Effect of dipterinyl calcium pentahydrate on hepatitis B virus replication in transgenic mice. J Transl Med 2010; 8:32. [PMID: 20356392 PMCID: PMC2853516 DOI: 10.1186/1479-5876-8-32] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Accepted: 03/31/2010] [Indexed: 12/26/2022] Open
Abstract
Background Dipterinyl calcium pentahydrate (DCP) has previously been shown to inhibit MDA-MB-231 human breast cancer xenographs in nude mice in a manner correlated with increases in plasma IL-12 and IL-4 concentrations, and decreases in plasma IL-6 levels. DCP also inhibits indoleamine 2,3-dioxygenase (IDO), an immuno-inhibitory enzyme, in human PBMCs (Peripheral Blood Mononuclear Cells). Methods In the present study, DCP was administered per os, once daily for 14 days to hepatitis B virus (HBV) transgenic mice at 23, 7.3, and 2.3 mg/(kg d). Multivariate stepwise regression and MANOVA analyses, by gender and treatment, of liver HBV DNA and RNA measures, liver core and serum HBe antigen assays, serum cytokine/chemokine profiles, and IDO metabolite measurements were performed. Results DCP caused a significant dose-response reduction of log liver HBV DNA as measured by PCR in the female HBV mice. The gender dependence of the anti-HBV DNA activity was explained by the DCP Effects Model (DCP-EM) (p = .001) which includes three serum biomarker changes caused by DCP: 1) decreased MCP-1; 2) decreased Kyn/Trp (an estimation of IDO activity); and 3) increased GM-CSF. Conclusions Immunomodulation via IDO or TDO (tryptophan 2,3-dioxygenase) pathways, along with serum MCP-1 and GM-CSF are proposed to play roles in the anti-HBV mechanism of DCP based upon their coordinated modulation in the reduction of viral DNA replication in HBV mice.
Collapse
Affiliation(s)
- Phillip Moheno
- SanRx Pharmaceuticals, Inc,, 603 Colima Street, La Jolla, California 92037-8032, USA.
| | | | | |
Collapse
|
45
|
Bhat A, Gomis S, Potter A, Tikoo SK. Role of Hsp90 in CpG ODN mediated immunostimulation in avian macrophages. Mol Immunol 2010; 47:1337-46. [PMID: 20096933 DOI: 10.1016/j.molimm.2009.12.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 12/23/2009] [Indexed: 12/29/2022]
Abstract
In mammals, CpG mediated immune activation is initiated through toll-like receptor (TLR) 9 and Hsp90 via activation of MAPK/ERK and PI3K/AKT pathways. However, in the absence of TLR9 ortholog in chicken genome, the role of Hsp90 and kinase (MAPK/ERK and PI3K/AKT) pathways in initiating CpG ODN(2007) induced immune activation in chicken is not clear. Using electrophoretic mobility shift assay (EMSA) and selective inhibitors of signal transduction pathways, we determined the role of these pathways in the production of Th1 cytokines/chemokines and nitric oxide (NO) in CpG ODN(2007) treated avian macrophage cells. Hsp90alpha but not Hsp90beta is bound to CpG ODN(2007). Inhibition of Hsp90 with geldanamycin resulted in the inactivation of MAPK/ERK and PI3K/AKT pathways leading to significantly reduced levels of IFN-gamma, IL-6 and NO mRNAs in CpG ODN(2007) stimulated cells. Moreover, inhibition of ERK1/2 and PI3/AKT kinase pathways with PD985009 and LY294002, respectively, suppresses the phosphorylation of ERK2 and AKT leading to the production of decreased amounts of IFN-gamma, IL-6 and NO mRNAs in CpG ODN(2007) stimulated cells. Our results demonstrate that binding of CpG ODN(2007) to Hsp90 induces activation of ERK2 and AKT phosphorylation leading to the production of high levels of IFN-gamma, IL-6, MIP-3alpha and nitric oxide (NO). In contrast to mammals, our results suggest that Hsp90alpha but not Hsp90beta binds with the CpG ODN(2007) and may play a major role in CpG ODN(2007) induced immunoactivation in avian macrophage cells. To our knowledge, this is the first report evaluating the involvement of Hsp90 and kinase (MAPK/ERK and PI3K/AKT) pathways in CpG mediated immunostimulation in avian macrophage cells.
Collapse
Affiliation(s)
- Audesh Bhat
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | | | | |
Collapse
|
46
|
Silveira FT, Lainson R, De Castro Gomes CM, Laurenti MD, Corbett CEP. Immunopathogenic competences of Leishmania (V.) braziliensis and L. (L.) amazonensis in American cutaneous leishmaniasis. Parasite Immunol 2009; 31:423-31. [PMID: 19646206 DOI: 10.1111/j.1365-3024.2009.01116.x] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The immunopathogenic competences of Leishmania (V.) braziliensis and L. (L.) amazonensis were reviewed in the light of more recent features found in the clinical and immunopathological spectrum of American cutaneous leishmaniasis. It was shown a dichotomy in the interaction between these Leishmania species and human T-cell immune response; while L. (V.) braziliensis shows a clear tendency to lead infection from the localized cutaneous leishmaniasis (LCL), a moderate T-cell hypersensitivity form at the centre of the spectrum, toward to the mucocutaneous leishmaniasis (MCL) at the T-cell hypersensitivity pole and with a prominent Th1-type immune response, L. (L.) amazonensis shows an opposite tendency, leading infection to the anergic diffuse cutaneous leishmaniasis (ADCL) at the T-cell hyposensitivity pole and with a marked Th2-type immune response. Between the central LCL and the two polar MCL and ADCL, the infection can present an intermediary form known as borderline disseminated cutaneous leishmaniasis, characterized by an incomplete inhibition of T-cell hypersensitivity but with a evident supremacy of Th1 over Th2 immune response (Th1 > or = Th2). These are probably the main immunopathogenic competences of L. (V.) braziliensis and L. (L.) amazonensis regarding the immune response dichotomy that modulates human infection outcome by these Leishmania parasites.
Collapse
Affiliation(s)
- F T Silveira
- Parasitology Department, Evandro Chagas Institute (Surveillance Secretary of Health, Ministry of Health), 66090-000, Belém, Pará State, Brazil.
| | | | | | | | | |
Collapse
|
47
|
Levesque MC, Hobbs MR, O'Loughlin CW, Chancellor JA, Chen Y, Tkachuk AN, Booth J, Patch KB, Allgood S, Pole AR, Fernandez CA, Mwaikambo ED, Mutabingwa TK, Fried M, Sorensen B, Duffy PE, Granger DL, Anstey NM, Weinberg JB. Malaria severity and human nitric oxide synthase type 2 (NOS2) promoter haplotypes. Hum Genet 2009; 127:163-82. [PMID: 19859740 DOI: 10.1007/s00439-009-0753-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 10/05/2009] [Indexed: 10/20/2022]
Abstract
Nitric oxide (NO) mediates host resistance to severe malaria and other infectious diseases. NO production and mononuclear cell expression of the NO producing enzyme-inducible nitric oxide synthase (NOS2) have been associated with protection from severe falciparum malaria. The purpose of this study was to identify single nucleotide polymorphisms (SNPs) and haplotypes in the NOS2 promoter, to identify associations of these haplotypes with malaria severity and to test the effects of these polymorphisms on promoter activity. We identified 34 SNPs in the proximal 7.3 kb region of the NOS2 promoter and inferred NOS2 promoter haplotypes based on genotyping 24 of these SNPs in a population of Tanzanian children with and without cerebral malaria. We identified 71 haplotypes; 24 of these haplotypes comprised 82% of the alleles. We determined whether NOS2 promoter haplotypes were associated with malaria severity in two groups of subjects from Dar es Salaam (N = 185 and N = 250) and in an inception cohort of children from Muheza-Tanga, Tanzania (N = 883). We did not find consistent associations of NOS2 promoter haplotypes with malaria severity or malarial anemia, although interpretation of these results was potentially limited by the sample size of each group. Furthermore, cytokine-induced NOS2 promoter activity determined using luciferase reporter constructs containing the proximal 7.3 kb region of the NOS2 promoter and the G-954C or C-1173T SNPs did not differ from NOS2 promoter constructs that lacked these polymorphisms. Taken together, these studies suggest that the relationship between NOS2 promoter polymorphisms and malaria severity is more complex than previously described.
Collapse
|
48
|
Weinberg JB, Chen Y, Jiang N, Beasley BE, Salerno JC, Ghosh DK. Inhibition of nitric oxide synthase by cobalamins and cobinamides. Free Radic Biol Med 2009; 46:1626-32. [PMID: 19328848 PMCID: PMC2745708 DOI: 10.1016/j.freeradbiomed.2009.03.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 03/18/2009] [Indexed: 11/22/2022]
Abstract
Cobalamins are important cofactors for methionine synthase and methylmalonyl-CoA mutase. Certain corrins also bind nitric oxide (NO), quenching its bioactivity. To determine if corrins would inhibit NO synthase (NOS), we measured their effects on -L-[(14)C]arginine-to-L-[(14)C]citrulline conversion by NOS1, NOS2, and NOS3. Hydroxocobalamin (OH-Cbl), cobinamide, and dicyanocobinamide (CN(2)-Cbi) potently inhibited all isoforms, whereas cyanocobalamin, methylcobalamin, and adenosylcobalamin had much less effect. OH-Cbl and CN(2)-Cbi prevented binding of the oxygen analog carbon monoxide (CO) to the reduced NOS1 and NOS2 heme active site. CN(2)-Cbi did not react directly with NO or CO. Spectral perturbation analysis showed that CN(2)-Cbi interacted directly with the purified NOS1 oxygenase domain. NOS inhibition by corrins was rapid and not reversed by dialysis with L-arginine or tetrahydrobiopterin. Molecular modeling indicated that corrins could access the unusually large heme- and substrate-binding pocket of NOS. Best fits were obtained in the "base-off" conformation of the lower axial dimethylbenzimidazole ligand. CN(2)-Cbi inhibited interferon-gamma-activated Raw264.7 mouse macrophage NO production. We show for the first time that certain corrins directly inhibit NOS, suggesting that these agents (or their derivatives) may have pharmacological utility. Endogenous cobalamins and cobinamides might play important roles in regulating NOS activity under normal and pathological conditions.
Collapse
Affiliation(s)
- J Brice Weinberg
- Division of Hematology-Oncology, Department of Medicine, Duke University and V.A. Medical Centers, Durham, NC 27705, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Kobzik L. Translating NO biology into clinical advances: still searching for the right dictionary? Am J Respir Cell Mol Biol 2009; 41:9-13. [PMID: 19448151 DOI: 10.1165/rcmb.2009-0156tr] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Lester Kobzik
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
50
|
Carsillo M, Kutala VK, Puschel K, Blanco J, Kuppusamy P, Niewiesk S. Nitric oxide production and nitric oxide synthase type 2 expression by cotton rat (Sigmodon hispidus) macrophages reflect the same pattern as human macrophages. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:718-24. [PMID: 19154756 PMCID: PMC2852107 DOI: 10.1016/j.dci.2008.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 12/18/2008] [Accepted: 12/20/2008] [Indexed: 05/27/2023]
Abstract
Our knowledge of the antibacterial role of nitric oxide (NO) during infection is based on studies of murine macrophages, which secrete large amounts of NO. In contrast, human macrophages produce very little NO and its relevance as an antibacterial mediator during infection of humans is uncertain. We have defined bone marrow-derived macrophages from cotton rats (Sigmodon hispidus). These macrophages display phenotypical and functional characteristics similar to other rodent and human macrophages. The most interesting finding was the low level of NO production which is in contrast to findings for murine macrophages, but consistent with those of humans. In spite of these low levels, inhibition of NO production led to a decrease in killing of bacteria. Cotton rats are highly susceptible to a variety of human pathogens and therefore offer a rodent model of infectious diseases with similar characteristics to humans in terms of NO production.
Collapse
Affiliation(s)
- Mary Carsillo
- Department of Veterinary Biosciences, The Ohio State University, OH, USA
| | | | - Karen Puschel
- Department of Veterinary Biosciences, The Ohio State University, OH, USA
| | | | | | - Stefan Niewiesk
- Department of Veterinary Biosciences, The Ohio State University, OH, USA
| |
Collapse
|