51
|
Li J, Duan H, Liu Y, Wang L, Zhou X. Biomaterial-Based Therapeutic Strategies for Obesity and Its Comorbidities. Pharmaceutics 2022; 14:1445. [PMID: 35890340 PMCID: PMC9320151 DOI: 10.3390/pharmaceutics14071445] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is a global public health issue that results in many health complications or comorbidities, including type 2 diabetes mellitus, cardiovascular disease, and fatty liver. Pharmacotherapy alone or combined with either lifestyle alteration or surgery represents the main modality to combat obesity and its complications. However, most anti-obesity drugs are limited by their bioavailability, target specificity, and potential toxic effects. Only a handful of drugs, including orlistat, liraglutide, and semaglutide, are currently approved for clinical obesity treatment. Thus, there is an urgent need for alternative treatment strategies. Based on the new revelation of the pathogenesis of obesity and the efforts toward the multi-disciplinary integration of materials, chemistry, biotechnology, and pharmacy, some emerging obesity treatment strategies are gradually entering the field of preclinical and clinical research. Herein, by analyzing the current situation and challenges of various new obesity treatment strategies such as small-molecule drugs, natural drugs, and biotechnology drugs, the advanced functions and prospects of biomaterials in obesity-targeted delivery, as well as their biological activities and applications in obesity treatment, are systematically summarized. Finally, based on the systematic analysis of biomaterial-based obesity therapeutic strategies, the future prospects and challenges in this field are proposed.
Collapse
Affiliation(s)
- Jing Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
| | - Hongli Duan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
| | - Yan Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
| | - Lu Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
| | - Xing Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (J.L.); (H.D.); (Y.L.)
- Institute of Materia Medica and Center of Translational Medicine, College of Pharmacy, Army Medical University, Chongqing 400038, China
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| |
Collapse
|
52
|
Liao KL, Watt KD. Mathematical Modeling and Analysis of CD200-CD200R in Cancer Treatment. Bull Math Biol 2022; 84:82. [PMID: 35792958 DOI: 10.1007/s11538-022-01039-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 06/01/2022] [Indexed: 11/26/2022]
Abstract
CD200 is a cell membrane protein that binds to its receptor, CD200 receptor (CD200R). The CD200 positive tumor cells inhibit the cellular functions of M1 and M2 macrophages and dendritic cells (DCs) through the CD200-CD200R complex, resulting in downregulation of Interleukin-10 and Interleukin-12 productions and affecting the activation of cytotoxic T lymphocytes. In this work, we provide two ordinary differential equation models, one complete model and one simplified model, to investigate how the binding affinities of CD200R and the populations of M1 and M2 macrophages affect the functions of the CD200-CD200R complex in tumor growth. Our simulations demonstrate that (i) the impact of the CD200-CD200R complex on tumor promotion or inhibition highly depends on the binding affinity of the CD200R on M2 macrophages and DCs to the CD200 on tumor cells, and (ii) a stronger binding affinity of the CD200R on M1 macrophages or DCs to the CD200 on tumor cells induces a higher tumor cell density in the CD200 positive tumor. Thus, the CD200 blockade would be an efficient treatment method in this case. Moreover, the simplified model shows that the binding affinity of CD200R on macrophages is the major factor to determine the treatment efficacy of CD200 blockade when the binding affinities of CD200R on M1 and M2 macrophages are significantly different to each other. On the other hand, both the binding affinity of CD200R and the population of macrophages are the major factors to determine the treatment efficacy of CD200 blockade when the binding affinities of CD200R on M1 and M2 macrophages are close to each other. We also analyze the simplified model to investigate the dynamics of the positive and trivial equilibria of the CD200 positive tumor case and the CD200 deficient tumor case. The bifurcation diagrams show that when M1 macrophages dominate the population, the tumor cell density of the CD200 positive tumor is higher than the one of CD200 deficient tumor. Moreover, the dynamics of tumor cell density change from tumor elimination to tumor persistence to oscillation, as the maximal proliferation rate of tumor cells increases.
Collapse
Affiliation(s)
- Kang-Ling Liao
- Department of Mathematics, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Kenton D Watt
- Department of Mathematics, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| |
Collapse
|
53
|
Diabetes Exacerbates Sepsis-Induced Neuroinflammation and Brain Mitochondrial Dysfunction. Inflammation 2022; 45:2352-2367. [PMID: 35689164 DOI: 10.1007/s10753-022-01697-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/06/2022] [Accepted: 06/01/2022] [Indexed: 11/05/2022]
Abstract
Sepsis is a life-threatening organ dysfunction, which demands notable attention for its treatment, especially in view of the involvement of immunodepressed patients, as the case of patients with diabetes mellitus (DM), who constitute a population susceptible to develop infections. Thus, considering this endocrine pathology as an implicatory role on the immune system, the aim of this study was to show the relationship between this disease and sepsis on neuroinflammatory and neurochemical parameters. Levels of IL-6, IL-10, brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and mitochondrial respiratory chain complexes were evaluated in the hippocampus and prefrontal cortex 24 h after sepsis by cecal ligation and perforation (CLP) in Wistar rats induced to type 1 diabetes by alloxan (150 mg/kg). It was verified that diabetes implied immune function after 24 h of sepsis, since it contributed to the increase of the inflammatory process with higher production of IL-6 and decreased levels of IL-10 only in the hippocampus. In the same brain area, a several decrease in NGF level and activity of complexes I and II of the mitochondrial respiratory chain were observed. Thus, diabetes exacerbates neuroinflammation and results in mitochondrial impairment and downregulation of NGF level in the hippocampus after sepsis.
Collapse
|
54
|
Yan Y, Xing C, Xiao Y, Shen X, Zhang Z, He C, Shi JB, Liu M, Liu X. Discovery and Anti-Inflammatory Activity Evaluation of a Novel CDK8 Inhibitor through Upregulation of IL-10 for the Treatment of Inflammatory Bowel Disease In Vivo. J Med Chem 2022; 65:7334-7362. [PMID: 35536548 DOI: 10.1021/acs.jmedchem.2c00356] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Increasing the anti-inflammatory cytokine interleukin-10 (IL-10) level is a promising strategy to suppress the progression of pathogenic inflammation including inflammatory bowel disease (IBD). Since cyclin-dependent kinase 8 (CDK8) inhibition can upregulate IL-10 abundance in activated myeloid-derived dendritic cells, it is considered to be an effective target for IBD treatment. Here, the complete discovery process of a novel CDK8 inhibitor as an anti-inflammatory agent was described. Starting with wogonin, structure-based optimization and structure-activity relationship (SAR) study were comprehensively carried out, and then lead compound 85 (N-(2-ethylphenyl)-5-(4-(piperazine-1-carbonyl)phenyl)nicotinamide) was developed as a potent druglike CDK8 inhibitor upregulating IL-10 both in vivo and in vitro. Also, compound 85 (with CDK8 IC50 = 56 nM, IL-10 enhancement rate 88%) exhibited effective anti-inflammatory activity in an animal model of IBD. These results confirmed that certain CDK8 inhibitor could be used as an effective anti-IBD drug.
Collapse
Affiliation(s)
- Yaoyao Yan
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, P. R. China
| | - Chen Xing
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, P. R. China
| | - Yun Xiao
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, P. R. China
| | - Xiaobao Shen
- School of Chemistry and Materials Engineering, Fuyang Normal University, Fuyang 236037, P. R. China
| | - Zhaoyan Zhang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, P. R. China
| | - Chuanbiao He
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, P. R. China
| | - Jing-Bo Shi
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, P. R. China
| | - Mingming Liu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, P. R. China
| | - Xinhua Liu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, P. R. China
| |
Collapse
|
55
|
Morris RM, Mortimer TO, O’Neill KL. Cytokines: Can Cancer Get the Message? Cancers (Basel) 2022; 14:cancers14092178. [PMID: 35565306 PMCID: PMC9103018 DOI: 10.3390/cancers14092178] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cytokines are important molecular players in cancer development, progression, and potential targets for treatment. Despite being small and overlooked, research has revealed that cytokines influence cancer biology in multiple ways. Cytokines are often found to contribute to immune function, cell damage, inflammation, angiogenesis, metastasis, and several other cellular processes important to tumor survival. Cytokines have also proven to have powerful effects on complex tumor microenvironment molecular biology and microbiology. Due to their heavy involvement in critical cancer-related processes, cytokines have also become attractive therapeutic targets for cancer treatment. In this review, we describe the relationship between several cytokines and crucial cancer-promoting processes and their therapeutic potential. Abstract Cytokines are small molecular messengers that have profound effects on cancer development. Increasing evidence shows that cytokines are heavily involved in regulating both pro- and antitumor activities, such as immune activation and suppression, inflammation, cell damage, angiogenesis, cancer stem-cell-like cell maintenance, invasion, and metastasis. Cytokines are often required to drive these cancer-related processes and, therefore, represent an important research area for understanding cancer development and the potential identification of novel therapeutic targets. Interestingly, some cytokines are reported to be related to both pro- and anti-tumorigenicity, indicating that cytokines may play several complex roles relating to cancer pathogenesis. In this review, we discuss some major cancer-related processes and their relationship with several cytokines.
Collapse
|
56
|
Lung injury induced by different negative suction pressure in patients with pneumoconiosis undergoing whole lung lavage. BMC Pulm Med 2022; 22:152. [PMID: 35459122 PMCID: PMC9034602 DOI: 10.1186/s12890-022-01952-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/15/2022] [Indexed: 11/10/2022] Open
Abstract
Background Pneumoconiosis is a diffuse interstitial fibronodular lung disease, which is caused by the inhalation of crystalline silica. Whole lung lavage (WLL) is a therapeutic procedure used to treat pneumoconiosis. This study is to compare the effects of different negative pressure suction on lung injury in patients with pneumoconiosis undergoing WLL. Materials and methods A prospective study was conducted with 24 consecutively pneumoconiosis patients who underwent WLL from March 2020 to July 2020 at Emergency General Hospital, China. The patients were divided into two groups: high negative suction pressure group (group H, n = 13, negative suction pressure of 300–400 mmHg) and low negative suction pressure group (group L, n = 11, negative suction pressure of 40–50 mmHg). The arterial blood gas, lung function, lavage data, oxidative stress, and inflammatory responses to access lung injury were monitored. Results Compared with those of group H, the right and left lung residual were significantly increased in the group L (P = 0.04, P = 0.01). Potential of hydrogen (pH), arterial partial pressure of oxygen (PaO2), arterial partial pressure of carbon dioxide (PaCO2), lactic acid (LAC) and glucose (GLU) varied from point to point in time (P < 0.01, respectively). There was statistical difference in the trend of superoxide dismutase (SOD) and interleukin-10 (IL-10) over time between the two groups (P < 0.01, P = 0.02). In comparison with the group H, the levels of IL-10 (P = 0.01) and SOD (P < 0.01) in WLL fluid were significantly increased in the group L. There was no statistical difference in the trend of maximal volumtary ventilation (MVV), forced vital capacity (FVC), forced expiratory volume in one second (FEV1%), residual volume (RV), residual volume/total lung capacity (RV/TLC), carbon monoxide dispersion factor (DLCO%), forced expiratory volume in one second/ forced vital capacity (FEV1/FVC%) over time between the two groups (P > 0.05, respectively). Conclusion Low negative suction pressure has the potential benefit to reduce lung injury in patients with pneumoconiosis undergoing WLL, although it can lead to increased residual lavage fluid. Despite differing suction strategies, pulmonary function parameters including FEV1%, RV and DLCO% became worse than before WLL. Trial Registration Chinese Clinical Trial registration number ChiCTR2000031024, 21/03/2020.
Collapse
|
57
|
Hsieh LS, Lu MS, Chiang WD. Identification and characterization of immunomodulatory peptides from pepsin-soy protein hydrolysates. BIORESOUR BIOPROCESS 2022; 9:39. [PMID: 38647785 PMCID: PMC10992351 DOI: 10.1186/s40643-022-00526-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
To obtain immunomodulatory peptides from isolated soy protein (ISP), pepsin was selected to prepare hydrolysates and 4-h treatment (Pepsin-ISPH4h) showed the highest yield and immunomodulatory activities. The Pepsin-ISPH4h was sequentially fractionated by 30, 10 and 1-kDa molecular weight cut-off (MWCO) membranes, in which 1-kDa MWCO permeate (1P) exhibited the most significant enhancement of phagocytosis activity without causing excessive inflammation as compared with Pepsin-ISPH4h. To further purify and enhance the immunomodulatory activity, 1P was distinct by high-performance liquid chromatography equipped with a reverse-phase column and in vivo immunomodulatory activity of fractions was examined in mice. Fraction 1 (F1) significantly elevated phagocytosis activity of mice spleen macrophages and neutrophils. However, increase of phagocytosis activity did not result from the induction of macrophages M1 or M2 polarization. The immunomodulatory peptide sequence, EKPQQQSSRRGS, from F1 was identified by LC-MS/MS. Phagocytosis activity and macrophage M1 polarization were elevated by synthetic peptide treatment. Hence, our results indicated that isolated soy protein hydrolysates prepared by pepsin could provide a source of peptides with immunomodulatory effects.
Collapse
Affiliation(s)
- Lu-Sheng Hsieh
- Department of Food Science, College of Agriculture, Tunghai University, No. 1727, Sec. 4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan
| | - Ming-Shing Lu
- Department of Food Science, College of Agriculture, Tunghai University, No. 1727, Sec. 4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan
| | - Wen-Dee Chiang
- Department of Food Science, College of Agriculture, Tunghai University, No. 1727, Sec. 4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan.
| |
Collapse
|
58
|
Czaja AJ. Immune Inhibitory Properties and Therapeutic Prospects of Transforming Growth Factor-Beta and Interleukin 10 in Autoimmune Hepatitis. Dig Dis Sci 2022; 67:1163-1186. [PMID: 33835375 DOI: 10.1007/s10620-021-06968-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/22/2021] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-beta and interleukin 10 have diverse immune inhibitory properties that have restored homeostatic defense mechanisms in experimental models of autoimmune disease. The goals of this review are to describe the actions of each cytokine, review their investigational use in animal models and patients, and indicate their prospects as interventions in autoimmune hepatitis. English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. Transforming growth factor-beta expands the natural and inducible populations of regulatory T cells, limits the proliferation of natural killer cells, suppresses the activation of naïve CD8+ T cells, decreases the production of interferon-gamma, and stimulates fibrotic repair. Interleukin 10 selectively inhibits the CD28 co-stimulatory signal for antigen recognition and impairs antigen-specific activation of uncommitted CD4+ and CD8+ T cells. It also inhibits maturation of dendritic cells, suppresses Th17 cells, supports regulatory T cells, and limits production of diverse pro-inflammatory cytokines. Contradictory immune stimulatory effects have been associated with each cytokine and may relate to the dose and accompanying cytokine milieu. Experimental findings have not translated into successful early clinical trials. The recombinant preparation of each agent in low dosage has been safe in human studies. In conclusion, transforming growth factor-beta and interleukin 10 have powerful immune inhibitory actions of potential therapeutic value in autoimmune hepatitis. The keys to their therapeutic application will be to match their predominant non-redundant function with the pivotal pathogenic mechanism or cytokine deficiency and to avoid contradictory immune stimulatory actions.
Collapse
Affiliation(s)
- Albert J Czaja
- Professor Emeritus of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
59
|
Rodrigues de Souza I, Savio de Araujo-Souza P, Morais Leme D. Genetic variants affecting chemical mediated skin immunotoxicity. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2022; 25:43-95. [PMID: 34979876 DOI: 10.1080/10937404.2021.2013372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The skin is an immune-competent organ and this function may be impaired by exposure to chemicals, which may ultimately result in immune-mediated dermal disorders. Interindividual variability to chemical-induced skin immune reactions is associated with intrinsic individual characteristics and their genomes. In the last 30-40 years, several genes influencing susceptibility to skin immune reactions were identified. The aim of this review is to provide information regarding common genetic variations affecting skin immunotoxicity. The polymorphisms selected for this review are related to xenobiotic-metabolizing enzymes (CYPA1 and CYPB1 genes), antioxidant defense (GSTM1, GSTT1, and GSTP1 genes), aryl hydrocarbon receptor signaling pathway (AHR and ARNT genes), skin barrier function transepidermal water loss (FLG, CASP14, and SPINK5 genes), inflammation (TNF, IL10, IL6, IL18, IL31, and TSLP genes), major histocompatibility complex (MHC) and neuroendocrine system peptides (CALCA, TRPV1, ACE genes). These genes present variants associated with skin immune responses and diseases, as well as variants associated with protecting skin immune homeostasis following chemical exposure. The molecular and association studies focusing on these genetic variants may elucidate their functional consequences and contribution in the susceptibility to skin immunotoxicity. Providing information on how genetic variations affect the skin immune system may reduce uncertainties in estimating chemical hazards/risks for human health in the future.
Collapse
Affiliation(s)
| | | | - Daniela Morais Leme
- Graduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Curitiba, Brazil
- National Institute for Alternative Technologies of Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactives (INCT-DATREM), Institute of Chemistry, Araraquara, Brazil
| |
Collapse
|
60
|
The use of supercytokines, immunocytokines, engager cytokines, and other synthetic cytokines in immunotherapy. Cell Mol Immunol 2022; 19:192-209. [PMID: 35043005 PMCID: PMC8803834 DOI: 10.1038/s41423-021-00786-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/25/2021] [Indexed: 02/08/2023] Open
Abstract
Cytokines exert powerful immunomodulatory effects that are critical to physiology and pathology in humans. The application of natural cytokines in clinical studies has not been clearly established, and there are often problems associated with toxicity or lack of efficacy. The key reasons can be attributed to the pleiotropy of cytokine receptors and undesired activation of off-target cells. With a deeper understanding of the structural principles and functional signals of cytokine-receptor interactions, artificial modification of cytokine signaling through protein engineering and synthetic immunology has become an increasingly feasible and powerful approach. Engineered cytokines are designed to selectively target cells. Herein, the theoretical and experimental evidence of cytokine engineering is reviewed, and the "supercytokines" resulting from structural enhancement and the "immunocytokines" generated by antibody fusion are described. Finally, the "engager cytokines" formed by the crosslinking of cytokines and bispecific immune engagers and other synthetic cytokines formed by nonnatural analogs are also discussed.
Collapse
|
61
|
Das SC, Rahman MA, Das Gupta S. In-silico analysis unravels the structural and functional consequences of non-synonymous SNPs in the human IL-10 gene. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00223-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Background
Interleukin-10 (IL-10) is an anti-inflammatory cytokine that affects different immune cells. It is also associated with the stimulation of the T and B cells for the production of antibodies. Several genetic polymorphisms in the IL-10 gene have been reported to cause or aggravate certain diseases like inflammatory bowel disease, rheumatoid arthritis, systemic sclerosis, asthma, etc. However, the disease susceptibility and abnormal function of the mutated IL-10 variants remain obscure.
Results
In this study, we used seven bioinformatics tools (SIFT, PROVEAN, PMut, PANTHER, PolyPhen-2, PHD-SNP, and SNPs&GO) to predict the disease susceptible non-synonymous SNPs (nsSNPs) of IL-10. Nine nsSNPs of IL-10 were predicted to be potentially deleterious: R42G, R45Q, F48L, E72G, M95T, A98D, R125S, Y155C, and I168T. Except two, all of the putative deleterious mutations are found in the highly conserved region of IL-10 protein structure, thus affecting the protein's stability. The 3-D structure of mutant proteins was modeled by project HOPE, and the protein–protein interactions were assessed with STRING. The predicted nsSNPs: R42Q, R45Q, F48L, E72G, and I168T are situated in the binding site region of the IL-10R1 receptor. Disruption of binding affinity with its receptor leads to deregulation of the JAK-STAT pathway and results in enhanced inflammation that imbalance in cellular signaling. Finally, Kaplan–Meier Plotter analysis displayed that deregulation of IL-10 expression affects gastric and ovarian cancer patients' survival rate. Thus, IL-10 could be useful as a potential prognostic marker gene for some cancers.
Conclusion
This study has determined the deleterious nsSNPs of IL-10 that might contribute to the malfunction of IL-10 protein and ultimately lead to the IL-10 associated diseases.
Collapse
|
62
|
Errera MH, Pratas A, Fisson S, Manicom T, Boubaya M, Sedira N, Héron E, Merabet L, Kobal A, Levy V, Warnet JM, Chaumeil C, Brignole-Baudouin F, Sahel JA, Goldschmidt P, Bodaghi B, Bloch-Queyrat C. Cytokines, chemokines and growth factors profile in human aqueous humor in idiopathic uveitis. PLoS One 2022; 17:e0254972. [PMID: 35061677 PMCID: PMC8782285 DOI: 10.1371/journal.pone.0254972] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
To investigate which cytokines, chemokines and growth factors are involved in the immunopathogenesis of idiopathic uveitis, and whether cytokine profiles are associated with. Serum and aqueous humor (AH) samples of 75 patients with idiopathic uveitis were analyzed by multiplex immunoassay. Infectious controls consisted of 16 patients with ocular toxoplasmosis all confirmed by intraocular fluid analyses. Noninfectious controls consisted of 7 patients with Behçet disease related uveitis and 15 patients with sarcoidosis related uveitis. The control group consisted of AH and serum samples from 47 noninflammatory control patients with age-related cataract. In each sample, 27 immune mediators ± IL-21 and IL-23 were measured. In idiopathic uveitis, 13 of the 29 mediators, including most proinflammatory and vascular mediators such as IL-6, IL-8, IL-12, G-CSF, GM-CSF, MCP-1, IP-10, TNF-α and VEGF, were significantly elevated in the aqueous humor when compared to all controls. Moreover, IL-17, IP-10, and IL-21, were significantly elevated in the serum when compared to all controls. We clustered 4 subgroups of idiopathic uveitis using a statistical analysis of hierarchical unsupervised classification, characterized by the order of magnitude of concentrations of intraocular cytokines. The pathogenesis of idiopathic uveitis is characterized by the presence of predominantly proinflammatory cytokines and chemokines and vascular endothelial growth factor with high expression levels as compared to other causes of uveitis. There are indications for obvious Th-1/ IL21-Th17 pathways but also IL9-Th9 and increased IFN-γ-inducing cytokine (IL12) and IFN-γ-inducible CXC chemokine (IP-10). The combined data suggest that immune mediator expression is different among idiopathic uveitis. This study suggests various clusters among the idiopathic uveitis group rather than one specific uveitis entity.
Collapse
Affiliation(s)
- Marie-Hélène Errera
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
- Sorbonne Universités, UPMC Univ Paris, Paris, France
- Department of Ophthalmology, UPMC Eye Center, University of Pittsburgh School of Medicine, Pennsylvania, United States of America
| | - Ana Pratas
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
| | - Sylvain Fisson
- Généthon, Inserm UMR_S951, Univ Evry, Université Paris-Saclay, EPHE, Evry, France
| | - Thomas Manicom
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
- Sorbonne Universités, UPMC Univ Paris, Paris, France
| | - Marouane Boubaya
- Université Paris 13, Sorbonne Paris cité, INSERM U1163/CNRS ERL 8254, AP-HP, Hôpital Avicenne, URC-CRC GHPSS, Bobigny, France
| | - Neila Sedira
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
| | - Emmanuel Héron
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
| | - Lilia Merabet
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
| | - Alfred Kobal
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
| | - Vincent Levy
- Université Paris 13, Sorbonne Paris cité, INSERM U1163/CNRS ERL 8254, AP-HP, Hôpital Avicenne, URC-CRC GHPSS, Bobigny, France
| | | | - Christine Chaumeil
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
| | - Françoise Brignole-Baudouin
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
- Faculty Pharmacy, Sorbonne Universities, Paris, France
| | - José-Alain Sahel
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
- Sorbonne Universités, UPMC Univ Paris, Paris, France
- Department of Ophthalmology, UPMC Eye Center, University of Pittsburgh School of Medicine, Pennsylvania, United States of America
| | - Pablo Goldschmidt
- Departments of Ophthalmology and Internal Medicine at Quinze-Vingts National Eye Hospital and DHU Sight Restore, Laboratory, Paris, France
| | - Bahram Bodaghi
- Sorbonne Universités, UPMC Univ Paris, Paris, France
- Pitié-Salpêtrière Hospital, DHU Sight Restore, Paris, France
| | - Coralie Bloch-Queyrat
- Université Paris 13, Sorbonne Paris cité, INSERM U1163/CNRS ERL 8254, AP-HP, Hôpital Avicenne, URC-CRC GHPSS, Bobigny, France
| |
Collapse
|
63
|
Abstract
Interleukins (ILs) and associated cytokines serve as the means of communication for immune cells and non-immune cells. The use of ILs in harnessing the immune system to cancer treatment has been a promising approach. ILs not only nurture an environment enabling cancer growth but also simultaneously trigger a productive tumor-directed immune response. These properties of ILs are increasingly being explored as a strategy to improve the outcomes of cancer. Here, we describe recently innovative technological approaches that have been developed to improve the pharmacokinetics, safety, and efficacies of IL-2, 15, 10, and 18 in the treatment of melanoma. Furthermore, the combination of ILs and immune checkpoint inhibition may synergize to reshape the tumor environment, thus yielding better clinical benefits in the future.
Collapse
|
64
|
Tamimou R, Lumbroso S, Mouzat K, Lopez-Castroman J. Genetic variations related to inflammation in suicidal ideation and behavior: A systematic review. Front Psychiatry 2022; 13:1003034. [PMID: 36325529 PMCID: PMC9621324 DOI: 10.3389/fpsyt.2022.1003034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND/OBJECTIVES Immune-inflammatory changes have been found in all types of suicidal ideation and behavior (SIB), independently of associated mental disorders. Since several Single Nucleotide Polymorphisms (SNPs) affect the function of inflammation-related genes, we searched the literature for genetic variations potentially altering inflammatory processes in SIB. METHODS We included studies that looked for associations between SIB and SNPs in genes related to inflammatory processes. Case reports, literature reviews, and animal studies were excluded. Articles were retrieved from PubMed and PsycINFO databases, Google Scholar and GreySource Index until September 17th, 2022. Quality was assessed using Q-Genie. RESULTS We analyzed 32 studies. SIB has been associated with eighteen SNPs located in genes encoding for interleukin-8 (rs4073), C-reactive protein (rs1130864), tumor necrosis factor α (rs1800629, rs361525, and rs1099724), tumor necrosis factor receptor 2 (rs1061622), transforming growth factor β-1 (rs1982073), acid phosphatase 1 (rs7419262, rs300774), interleukin-10 (rs1800896), interferon γ (rs2430561), amino-carboxy muconate semialdehyde decarboxylase (rs2121337), interleukin 7 (rs10448044, rs10448042), macrophage migration inhibitory factor (rs755622), interleukin 1-α (rs1800587), and interleukin 1-β (rs1143634 and rs16944. A genome-wide association study reported one association at the threshold of significance with the rs300774 SNP, located in the 2p25 region containing ACP1 gene. DISCUSSION The studies included were methodologically and clinically diverse and of moderate quality. Their findings suggest that some inflammation-related SNPs could increase the likelihood of SIB but the evidence to date is insufficient. Further research using gene-gene (GxG) and gene-environment (GxE) approaches is warranted. SYSTEMATIC REVIEW REGISTRATION [https://www.crd.york.ac.uk], identifier [CRD42022296310].
Collapse
Affiliation(s)
- Rabah Tamimou
- Department of Psychiatry, Nimes University Hospital, Nimes, France.,Laboratory of Biochemistry and Molecular Biology, Nimes University Hospital, University of Montpellier, Nimes, France.,Institut de Génomique Fonctionnelle, University of Montpellier, CNRS-INSERM, Montpellier, France
| | - Serge Lumbroso
- Laboratory of Biochemistry and Molecular Biology, Nimes University Hospital, University of Montpellier, Nimes, France
| | - Kevin Mouzat
- Laboratory of Biochemistry and Molecular Biology, Nimes University Hospital, University of Montpellier, Nimes, France
| | - Jorge Lopez-Castroman
- Department of Psychiatry, Nimes University Hospital, Nimes, France.,Institut de Génomique Fonctionnelle, University of Montpellier, CNRS-INSERM, Montpellier, France.,Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain
| |
Collapse
|
65
|
Huang Y, Zhai X, Ma T, Zhang M, Pan H, Weijia Lu W, Zhao X, Sun T, Li Y, Shen J, Yan C, Du Y. Rare earth-based materials for bone regeneration: Breakthroughs and advantages. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
66
|
Al-Khrashi LA, Badr AM, Al-Amin MA, Mahran YF. Thymol ameliorates 5-fluorouracil-induced intestinal mucositis: Evidence of down-regulatory effect on TGF-β/MAPK pathways through NF-κB. J Biochem Mol Toxicol 2022; 36:e22932. [PMID: 34665902 DOI: 10.1002/jbt.22932] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/31/2021] [Accepted: 10/06/2021] [Indexed: 12/11/2022]
Abstract
5-Fluorouracil (5-FU) is a front-line cytotoxic therapy. However, intestinal mucositis is a well-known adverse event of 5-FU, which limits its therapeutic use. Indeed, thymol, which is a monoterpene component of the essential oil derived from thymus, has a potential anti-inflammatory and immunomodulatory activity. Therefore, this study aimed to investigate the potential chemoprotective effect of thymol against 5-FU-induced intestinal mucositis. Rats were either exposed to two doses of 5-FU (150 mg/kg, ip) and/or treated with thymol (60 or 120 mg/kg). Oxidative stress and inflammatory markers, as well as pathological changes, were assessed. 5-FU-induced severe intestinal damages as were evidenced by histopathological changes as well as oxidative and inflammatory responses. Thymol pretreatment inhibited 5-FU-induced oxidative stress by reducing lipid peroxidation and increasing intestinal levels of antioxidant systems. Moreover, inflammatory response markers, such as interleukin-6, prostaglandin E2, and COX-2 were also improved. The immunoblotting analysis also showed that thymol significantly inhibited the 5-FU-induced expression of nuclear factor-κB, tumor necrosis factor-α, and transforming growth factor β-1 (TGF-β1), in addition to the suppression of p38 and phosphorylated c-Jun N-terminal kinases (p-JNK) mitogen-activated protein kinase proteins' expressions. Our study is the first to demonstrate the promising protective effect of thymol against 5-FU-induced intestinal mucositis through inhibition of oxidative, inflammatory pathways, and suppression of TGF-β/p38/p-JNK signaling.
Collapse
Affiliation(s)
- Layla A Al-Khrashi
- Department of Pharmacology and Toxicology, College of Pharmacy, KSU, Riyadh, Saudi Arabia
| | - Amira M Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, KSU, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Maha A Al-Amin
- Department of Pharmacology and Toxicology, College of Pharmacy, KSU, Riyadh, Saudi Arabia
| | - Yasmen F Mahran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
67
|
Amaroli A, Pasquale C, Zekiy A, Benedicenti S, Marchegiani A, Sabbieti MG, Agas D. Steering the multipotent mesenchymal cells towards an anti-inflammatory and osteogenic bias via photobiomodulation therapy: How to kill two birds with one stone. J Tissue Eng 2022; 13:20417314221110192. [PMID: 35832724 PMCID: PMC9272199 DOI: 10.1177/20417314221110192] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/13/2022] [Indexed: 12/17/2022] Open
Abstract
The bone marrow-derived multipotent mesenchymal cells (MSCs) have captured scientific interest due to their multi-purpose features and clinical applications. The operational dimension of MSCs is not limited to the bone marrow reservoir, which exerts bone-building and niche anabolic tasks; they also meet the needs of quenching inflammation and restoring inflamed tissues. Thus, the range of MSC activities extends to conditions such as neurodegenerative diseases, immune disorders and various forms of osteopenia. Steering these cells towards becoming an effective therapeutic tool has become mandatory. Many laboratories have employed distinct strategies to improve the plasticity and secretome of MSCs. We aimed to present how photobiomodulation therapy (PBM-t) can manipulate MSCs to render them an extraordinary anti-inflammatory and osteogenic instrument. Moreover, we discuss the outcomes of different PBM-t protocols on MSCs, concluding with some perplexities and complexities of PBM-t in vivo but encouraging and feasible in vitro solutions.
Collapse
Affiliation(s)
- Andrea Amaroli
- Department of Surgical and Diagnostic Sciences, University of Genoa, Genoa, Italy.,Department of Orthopedic Dentistry, Faculty of Dentistry, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Claudio Pasquale
- Department of Surgical and Diagnostic Sciences, University of Genoa, Genoa, Italy
| | - Angelina Zekiy
- Department of Orthopedic Dentistry, Faculty of Dentistry, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Stefano Benedicenti
- Department of Surgical and Diagnostic Sciences, University of Genoa, Genoa, Italy
| | - Andrea Marchegiani
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | | | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| |
Collapse
|
68
|
Toita R, Kawano T, Murata M, Kang JH. Bioinspired macrophage-targeted anti-inflammatory nanomedicine: A therapeutic option for the treatment of myocarditis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112492. [PMID: 34857278 DOI: 10.1016/j.msec.2021.112492] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/29/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Myocarditis is a disease characterized by inflammation of the heart muscle, which increases the risk of dilated cardiomyopathy and heart failure. Macrophage migration is a major histopathological hallmark of myocarditis, making macrophages a potential therapeutic target for the management of this disease. In the present study, we synthesized a bioinspired anti-inflammatory nanomedicine conjugated with protein G (PSL-G) that could target macrophages and induce macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. Notably, PSL-G exhibited a higher affinity for macrophages than non-macrophage cells. The addition of PSL-G decreased the levels of pro-inflammatory cytokines (e.g., IL-1α, IL-6, and TNF-α), but increased the level of the anti-inflammatory cytokine IL-10 in macrophages treated with lipopolysaccharide and/or interferon-γ. Furthermore, the lifetime of PSL-G in murine blood circulation was found to be significantly higher than that of PSL. Systemic injection of PSL-G into a mouse model of experimental autoimmune myocarditis remarkably reduced macrophage migration in the myocardium (16-fold compared with the positive control group) and myocardial fibrosis (8-fold). Based on these results and the fact that macrophages play a critical role in the pathogenesis of various diseases, we believe that bioinspired macrophage-targeted anti-inflammatory nanomedicines may be effective therapeutic options for the treatment of autoimmune and autoinflammatory diseases, especially myocarditis.
Collapse
Affiliation(s)
- Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan; AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jeong-Hun Kang
- Division of Biopharmaceutics and Pharmacokinetics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan.
| |
Collapse
|
69
|
ROS-Based Nanoparticles for Atherosclerosis Treatment. MATERIALS 2021; 14:ma14226921. [PMID: 34832328 PMCID: PMC8619986 DOI: 10.3390/ma14226921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/29/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
Atherosclerosis (AS), a chronic arterial disease, is the leading cause of death in western developed countries. Considering its long-term asymptomatic progression and serious complications, the early prevention and effective treatment of AS are particularly important. The unique characteristics of nanoparticles (NPs) make them attractive in novel therapeutic and diagnostic applications, providing new options for the treatment of AS. With the assistance of reactive oxygen species (ROS)-based NPs, drugs can reach specific lesion areas, prolong the therapeutic effect, achieve targeted controlled release and reduce adverse side effects. In this article, we reviewed the mechanism of AS and the generation and removal strategy of ROS. We further discussed ROS-based NPs, and summarized their biomedical applications in scavenger and drug delivery. Furthermore, we highlighted the recent advances, challenges and future perspectives of ROS-based NPs for treating AS.
Collapse
|
70
|
Rafaqat S, Rafaqat S, Rafaqat S. Pathophysiological role of major adipokines in Atrial Fibrillation. INTERNATIONAL JOURNAL OF ARRHYTHMIA 2021. [DOI: 10.1186/s42444-021-00048-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The adipokines, secreted from adipose tissue or body fats, are also called adipocytokines which are cytokines, cell signaling proteins or cell–cell communication. However, AF is a common cardiac arrhythmia in which the heart beats so fast by abnormal beating and is a serious public health disease associated with increased heart failure, systemic thromboembolism, and death. Adipokines are cardiovascular disease (CVD) mediators or biomarkers that affect the heart as well as blood vessels, by increasing the cardiac contractility and action potential duration, which result in the extent of left ventricular and atrial remodeling.
Main body
Google Scholar, PubMed, and science direct were used to review the literature. Many keywords were used for searching the literature such as Adipokines, Leptin, Apelin, Adiponectin, Omentin-1, Chemerin, CTRP3, TNF-α, IL-6, IL-10, and AF. According to the literature, much more data are available for numerous adipokines, but this review article only has taken few major adipokines which played their major role in Atrial Fibrillation. The review article did not limit the time frame.
Conclusion
In conclusion, adipokines play a significant role in the development and progress of atrial fibrillation. Also, there are major adipokines such as adiponectin, apelin, C1q/TNF-Related Protein 3 (CTRP3), Chemerin, Omentin-1, interleukin-6, Leptin, TNF-α, resistin, and interleukin-10, which played their pathophysiological role in atrial fibrillation by causing cardiac hypertrophy, increasing the cardiac contractility and action potential duration, atrial fibrosis, electrical and structural remodeling of atrial tissue.
Collapse
|
71
|
Zhang J, Zhao H, Feng Y, Xu X, Yang Y, Zhang P, Lu Z, Zhang T. Topoisomerase 2 inhibitor etoposide promotes interleukin-10 production in LPS-induced macrophages via upregulating transcription factor Maf and activating PI3K/Akt pathway. Int Immunopharmacol 2021; 101:108264. [PMID: 34715493 DOI: 10.1016/j.intimp.2021.108264] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 01/21/2023]
Abstract
Topoisomerase (TOP) inhibitors were commonly used as chemotherapeutic agents in the treatment of cancers. In our present study, we found that etoposide (ETO), a topoisomerase 2 (TOP2) inhibitor, upregulated the production of Interleukin 10 (IL-10) in lipopolysaccharide (LPS)-stimulated macrophages. Besides, other TOP2 inhibitors including doxorubicin hydrochloride (DOX) and teniposide (TEN) were also able to augment IL-10 production. Meanwhile, the expression levels of pro-inflammatory factors, for example IL-6 and TNF-α, were also decreased accordingly by the treatment of the TOP2 inhibitors. Of note, ETO facilitated IL-10 secretion, which might be regulated by transcription factor Maf via PI3K/AKT pathway, as pharmaceutic blockage of kinase PI3K or AKT attenuated ETO-induced Maf and IL-10 expression. Further, in LPS-induced mice sepsis model, the enhanced generation of IL-10 was observed in ETO-treated mice, whereas pro-inflammatory cytokines were decreased, which significantly reduced the mortality of mice from LPS-induced lethal cytokine storm. Taken together, these results indicated that ETO may exhibit an anti-inflammatory role by upregulating the alteration of transcription factor Maf and promoting subsequential IL-10 secretion via PI3K/Akt pathway in LPS-induced macrophages. Therefore, ETO may serve as a potential anti-inflammatory agent and employed to severe pro-inflammatory diseases including COVID-19.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Department of Immunology, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, China; Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China; Key Laboratory of Microecology-immune Regulatory Network and Related Diseases of Heilongjiang Province, Jiamusi, Heilongjiang, China
| | - Haoxin Zhao
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Yuan Feng
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China; Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Xin Xu
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Yili Yang
- China Regional Research Center, International Centre for Genetic Engineering and Biotechnology, Taizhou, China
| | - Pengxia Zhang
- Department of Immunology, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, China; Key Laboratory of Microecology-immune Regulatory Network and Related Diseases of Heilongjiang Province, Jiamusi, Heilongjiang, China.
| | - Zhiliang Lu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.
| | - Tao Zhang
- Department of Immunology, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, China; Key Laboratory of Microecology-immune Regulatory Network and Related Diseases of Heilongjiang Province, Jiamusi, Heilongjiang, China.
| |
Collapse
|
72
|
Souza MRD, Hilário Garcia AL, Dalberto D, Martins G, Picinini J, Souza GMSD, Chytry P, Dias JF, Bobermin LD, Quincozes-Santos A, da Silva J. Environmental exposure to mineral coal and by-products: Influence on human health and genomic instability. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117346. [PMID: 34020260 DOI: 10.1016/j.envpol.2021.117346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 06/12/2023]
Abstract
Environmental exposure to pollution generated by mining and burning coal is inevitable for people living nearby. Therefore, the aim of this study was to evaluate the influence of coal dust on health conditions and genomic instability of individuals who live near coal mines and thermoelectric power plants, and to relate the results to inorganic elements and inflammatory responses. Thus, we evaluated 284 individuals from four cities in the south of Brazil around a region with coal mines and a thermoelectric power plant (one city was considered a negative control). The results of the Comet assay and Micronucleus (MN) test did not show a genotoxic or mutagenic effect related to environmental exposure to coal, but the inflammatory cytokine tumor necrosis factor-α (TNF-α) was increased in all cities around the power plant when compared to the control conditions. Higher levels of MN were associated with body mass index and cardiovascular risk, and higher levels of Damage Index (DI), TNF-α and interleukin1β (IL-1β) with number of cigarettes/day. Principal component analysis (PCA) was used to integrate DNA damage and inflammatory results with inorganic elements. This study also demonstrated the relationship between zinc and MN, copper, and interleukin10 (IL-10), and among silicon and sulfur with DI and nucleoplasmic bridge. A relationship was also observed between the reduction of inorganic elements and both aging and quality of health. The use of different methodologies and the relationship between the results obtained in these studies, including different lifestyles, can increase the understanding of the interaction between this mineral and the health status of residents of regions affected by coal pollution.
Collapse
Affiliation(s)
- Melissa Rosa de Souza
- Laboratory of Genetic Toxicology, Postgraduate Program in Molecular and Cell Biology Applied to Health, Lutheran University of Brazil (ULBRA), Avenida Farroupilha, 8001 Bairro São José, CEP 92425-900, Canoas, RS, Brazil.
| | - Ana Letícia Hilário Garcia
- Laboratory of Genetic Toxicology, Postgraduate Program in Molecular and Cell Biology Applied to Health, Lutheran University of Brazil (ULBRA), Avenida Farroupilha, 8001 Bairro São José, CEP 92425-900, Canoas, RS, Brazil
| | - Daiana Dalberto
- Laboratory of Genetic Toxicology, Postgraduate Program in Molecular and Cell Biology Applied to Health, Lutheran University of Brazil (ULBRA), Avenida Farroupilha, 8001 Bairro São José, CEP 92425-900, Canoas, RS, Brazil
| | - Gabriela Martins
- Laboratory of Genetic Toxicology, Postgraduate Program in Molecular and Cell Biology Applied to Health, Lutheran University of Brazil (ULBRA), Avenida Farroupilha, 8001 Bairro São José, CEP 92425-900, Canoas, RS, Brazil
| | - Juliana Picinini
- Laboratory of Genetic Toxicology, Postgraduate Program in Molecular and Cell Biology Applied to Health, Lutheran University of Brazil (ULBRA), Avenida Farroupilha, 8001 Bairro São José, CEP 92425-900, Canoas, RS, Brazil
| | | | - Paola Chytry
- Ion Implantation Laboratory, Institute of Physics, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Johnny Ferraz Dias
- Ion Implantation Laboratory, Institute of Physics, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - André Quincozes-Santos
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Juliana da Silva
- Laboratory of Genetic Toxicology, Postgraduate Program in Molecular and Cell Biology Applied to Health, Lutheran University of Brazil (ULBRA), Avenida Farroupilha, 8001 Bairro São José, CEP 92425-900, Canoas, RS, Brazil
| |
Collapse
|
73
|
Micheli L, Durante M, Lucarini E, Sgambellone S, Lucarini L, Di Cesare Mannelli L, Ghelardini C, Masini E. The Histamine H 4 Receptor Participates in the Anti-Neuropathic Effect of the Adenosine A 3 Receptor Agonist IB-MECA: Role of CD4 + T Cells. Biomolecules 2021; 11:biom11101447. [PMID: 34680083 PMCID: PMC8533073 DOI: 10.3390/biom11101447] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/17/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
A3 adenosine receptor (A3AR) agonists have emerged as potent relievers of neuropathic pain by a T cell-mediated production of IL-10. The H4 histamine receptor (H4R), also implicated in pain modulation, is expressed on T cells playing a preeminent role in its activation and release of IL-10. To improve the therapeutic opportunities, this study aimed to verify the hypothesis of a possible cross-talk between A3AR and H4R in the resolution of neuropathic pain. In the mouse model of Chronic Constriction Injury (CCI), the acute intraperitoneal co-administration of the A3AR agonist IB-MECA (0.5 mg/kg) and the H4R agonist VUF 8430 (10 mg/kg), were additive in counteracting mechano-allodynia increasing IL-10 plasma levels. In H4R−/− mice, IB-MECA activity was reduced, lower pain relief and lower modulation of plasma IL-1β, TNF-α, IL-6 and IL-10 were shown. The complete anti-allodynia effect of IB-MECA in H4R−/− mice was restored after intravenous administration of CD4+ T cells obtained from naïve wild type mice. In conclusion, a role of the histaminergic system in the mechanism of A3AR-mediated neuropathic pain relief was suggested highlighting the driving force evoked by CD4+ T cells throughout IL-10 up-regulation.
Collapse
|
74
|
Cytokines and apoptosis in atopic dermatitis. Postepy Dermatol Alergol 2021; 38:1-13. [PMID: 34408560 PMCID: PMC8362769 DOI: 10.5114/ada.2019.88394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/29/2019] [Indexed: 02/06/2023] Open
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease. AD affects 10-20% of children worldwide and persists into adulthood in a minority of cases, affecting approximately 2-3% of the adult population, with an increased prevalence over the past decades in developed countries. Atopy is a genetic tendency to overproduce IgE class antibodies in response to common antigens found in the environment. Concurrence of different atopy such as allergic rhinitis or asthma in children with AD is estimated at 80%. AD is characterized by a vicious cycle of an allergic immune response. The emerging picture of the AD is a complex disorder with barrier dysfunction, immunological, genetic and environmental factors all playing key roles. Patients with severe or persistent disease and their families experience significant impairment in their quality of life, and in addition, AD places a heavy economic burden on society as a whole. Pathogenesis, the role of the epidermal barrier, mechanisms of cells apoptosis, the role of T cells and cytokines in AD are discussed in this article.
Collapse
|
75
|
Uricoli B, Birnbaum LA, Do P, Kelvin JM, Jain J, Costanza E, Chyong A, Porter CC, Rafiq S, Dreaden EC. Engineered Cytokines for Cancer and Autoimmune Disease Immunotherapy. Adv Healthc Mater 2021; 10:e2002214. [PMID: 33690997 PMCID: PMC8651077 DOI: 10.1002/adhm.202002214] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/15/2021] [Indexed: 12/17/2022]
Abstract
Cytokine signaling is critical to a range of biological processes including cell development, tissue repair, aging, and immunity. In addition to acting as key signal mediators of the immune system, cytokines can also serve as potent immunotherapies with more than 20 recombinant products currently Food and Drug Administration (FDA)-approved to treat conditions including hepatitis, multiple sclerosis, arthritis, and various cancers. Yet despite their biological importance and clinical utility, cytokine immunotherapies suffer from intrinsic challenges that limit their therapeutic potential including poor circulation, systemic toxicity, and low tissue- or cell-specificity. In the past decade in particular, methods have been devised to engineer cytokines in order to overcome such challenges and here, the myriad strategies are reviewed that may be employed in order to improve the therapeutic potential of cytokine and chemokine immunotherapies with applications in cancer and autoimmune disease therapy, as well as tissue engineering and regenerative medicine. For clarity, these strategies are collected and presented as they vary across size scales, ranging from single amino acid substitutions, to larger protein-polymer conjugates, nano/micrometer-scale particles, and macroscale implants. Together, this work aims to provide readers with a timely view of the field of cytokine engineering with an emphasis on early-stage therapeutic approaches.
Collapse
Affiliation(s)
- Biaggio Uricoli
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Lacey A. Birnbaum
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Priscilla Do
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - James M. Kelvin
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Juhi Jain
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory School of Medicine, Atlanta, GA 30322, USA
| | - Emma Costanza
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Andrew Chyong
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Christopher C. Porter
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology at Emory University School of Medicine
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Erik C. Dreaden
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
76
|
Pires IS, Hammond PT, Irvine DJ. Engineering Strategies for Immunomodulatory Cytokine Therapies - Challenges and Clinical Progress. ADVANCED THERAPEUTICS 2021; 4:2100035. [PMID: 34734110 PMCID: PMC8562465 DOI: 10.1002/adtp.202100035] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Indexed: 12/15/2022]
Abstract
Cytokines are immunoregulatory proteins involved in many pathological states with promising potential as therapeutic agents. A diverse array of cytokines have been studied in preclinical disease models since the 1950s, some of which became successful biopharmaceutical products with the advancement of recombinant protein technology in the 1980s. However, following these early approvals, clinical translation of these natural immune signaling molecules has been limited due to their pleiotropic action in many cell types, and the fact that they have evolved to act primarily locally in tissues. These characteristics, combined with poor pharmacokinetics, have hindered the delivery of cytokines via systemic administration routes due to dose-limiting toxicities. However, given their clinical potential and recent clinical successes in cancer immunotherapy, cytokines continue to be extensively pursued in preclinical and clinical studies, and a range of molecular and formulation engineering strategies are being applied to reduce treatment toxicity while maintaining or enhancing therapeutic efficacy. This review provides a brief background on the characteristics of cytokines and their history as clinical therapeutics, followed by a deeper discussion on the engineering strategies developed for cytokine therapies with a focus on the translational relevance of these approaches.
Collapse
Affiliation(s)
- Ivan S Pires
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
77
|
Hot or cold: Bioengineering immune contextures into in vitro patient-derived tumor models. Adv Drug Deliv Rev 2021; 175:113791. [PMID: 33965462 DOI: 10.1016/j.addr.2021.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have proven to be tremendously effective for a subset of cancer patients. However, it is difficult to predict the response of individual patients and efforts are now directed at understanding the mechanisms of ICI resistance. Current models of patient tumors poorly recapitulate the immune contexture, which describe immune parameters that are associated with patient survival. In this Review, we discuss parameters that influence the induction of different immune contextures found within tumors and how engineering strategies may be leveraged to recapitulate these contextures to develop the next generation of immune-competent patient-derived in vitro models.
Collapse
|
78
|
Moll-Bernardes R, de Sousa AS, Macedo AVS, Lopes RD, Vera N, Maia LCR, Feldman A, Arruda GDAS, Castro MJC, Pimentel-Coelho PM, de Albuquerque DC, de Paula TC, Furquim TAB, Loures VA, Giusti KGD, de Oliveira NM, De Luca FA, Kotsugai MDM, Domiciano RAM, Santos MF, de Souza OF, Bozza FA, Luiz RR, Medei E. IL-10 and IL-12 (P70) Levels Predict the Risk of Covid-19 Progression in Hypertensive Patients: Insights From the BRACE-CORONA Trial. Front Cardiovasc Med 2021; 8:702507. [PMID: 34386533 PMCID: PMC8353079 DOI: 10.3389/fcvm.2021.702507] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/02/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Cardiovascular comorbidities such as hypertension and inflammatory response dysregulation are associated with worse COVID-19 prognoses. Different cytokines have been proposed to play vital pathophysiological roles in COVID-19 progression, but appropriate prognostic biomarkers remain lacking. We hypothesized that the combination of immunological and clinical variables at admission could predict the clinical progression of COVID-19 in hypertensive patients. Methods: The levels of biomarkers, including C-reactive protein, lymphocytes, monocytes, and a panel of 29 cytokines, were measured in blood samples from 167 hypertensive patients included in the BRACE-CORONA trial. The primary outcome was the highest score during hospitalization on the modified WHO Ordinal Scale for Clinical Improvement. The probability of progression to severe disease was estimated using a logistic regression model that included clinical variables and biomarkers associated significantly with the primary outcome. Results: During hospitalization, 13 (7.8%) patients showed progression to more severe forms of COVID-19, including three deaths. Obesity, diabetes, oxygen saturation, lung involvement on computed tomography examination, the C-reactive protein level, levels of 15 cytokines, and lymphopenia on admission were associated with progression to severe COVID-19. Elevated levels of interleukin-10 and interleukin-12 (p70) combined with two or three of the abovementioned clinical comorbidities were associated strongly with progression to severe COVID-19. The risk of progression to severe disease reached 97.5% in the presence of the five variables included in our model. Conclusions: This study demonstrated that interleukin-10 and interleukin-12 (p70) levels, in combination with clinical variables, at hospital admission are key biomarkers associated with an increased risk of disease progression in hypertensive patients with COVID-19.
Collapse
Affiliation(s)
| | - Andrea Silvestre de Sousa
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Evandro Chagas National Institute of Infectious Disease, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ariane V S Macedo
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Hospital São Luiz Jabaquara, São Paulo, Brazil.,Santa Casa de São Paulo, São Paulo, Brazil
| | - Renato D Lopes
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, United States.,Brazilian Clinical Research Institute, São Paulo, Brazil
| | - Narendra Vera
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana C R Maia
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Feldman
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Hospital São Luiz Anália Franco, São Paulo, Brazil
| | | | - Mauro J C Castro
- Instituto de Microbiologia Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro M Pimentel-Coelho
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denílson C de Albuquerque
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Cardiology Department, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | | - Olga Ferreira de Souza
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Hospital Copa Star, Rio de Janeiro, Brazil
| | - Fernando A Bozza
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Evandro Chagas National Institute of Infectious Disease, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ronir Raggio Luiz
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Institute for Studies in Public Health-IESC, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emiliano Medei
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
79
|
Mansour NM, Elkalla WS, Ragab YM, Ramadan MA. Inhibition of acetic acid-induced colitis in rats by new Pediococcus acidilactici strains, vitamin producers recovered from human gut microbiota. PLoS One 2021; 16:e0255092. [PMID: 34310635 PMCID: PMC8312973 DOI: 10.1371/journal.pone.0255092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/09/2021] [Indexed: 01/24/2023] Open
Abstract
Our aim was to isolate, identify and characterize probiotic bacteria as vitamin producers in particular B2 and B9. 150 human fecal samples were collected and used for isolation of vitamin producers—probiotics. 49 isolates were chosen for screening their genome by PCR for the presence of riboflavin and folic acid genes. As a result, three isolates were selected and their production of the B2 and B9 were confirmed by HPLC. The three isolates were identified on species level by sequencing their 16S rRNA gene which showed 100% identical to strains of Pediococcus acidilactici. Thus, they were named as P. acidilactici WNYM01, P. acidilactici WNYM02, P. acidilactici WNYM03 and submitted to the Genbank database with accession numbers. They met the probiotic criteria by expressing 90–95% survival rate at pH (2.0–9.0) and bile salt up to 2% for 3 h in addition to their antimicrobial activity against gram positive and negative microorganisms. They also showed no hemolytic activity and common pattern for antibiotic susceptibility. Our three strains were tested individually or in mixture in vivo on rat colitis model compared to ulcerative group. The strains were administrated orally to rats in daily dose containing CFU 109 for 14 days then followed by induction of colitis using acetic acid then the oral administration was continued for more four days. The histology results, the anti-inflammatory and anti-oxidative stress biomarkers showed the protective role of the strains compared to the ulcerative group. As a conclusion, we introduce novel three probiotic candidates for pharmaceutical preparations and health applications.
Collapse
Affiliation(s)
- Nahla M. Mansour
- Gut Microbiology & Immunology Group, Chemistry of Natural and Microbial Products Dept., Pharmaceutical Industries Div., National Research Centre, Cairo, Egypt
- * E-mail:
| | - Wagiha S. Elkalla
- Microbiology and Immunology Department, Faculty of Pharmacy, Badr University in Cairo, Cairo, Egypt
| | - Yasser M. Ragab
- Microbiology and Immunology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohamed A. Ramadan
- Microbiology and Immunology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
80
|
Li J, Ding F, Qian X, Sun J, Ge Z, Yang L, Cheng Z. Anti-inflammatory cytokine IL10 loaded cRGD liposomes for the targeted treatment of atherosclerosis. J Microencapsul 2021; 38:357-364. [PMID: 33048003 DOI: 10.1080/02652048.2020.1836058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AIM Atherosclerosis (AS) is one of the main causes of cardiovascular disease which might lead to myocardial infarction or stroke and further leads to fatality. METHOD In this study, we have designed an anti-inflammatory cytokine interleukin-10 (IL10) delivery system to effectively alleviate the inflammation of atherosclerosis plaque. The targeted delivery of IL10 to the atherosclerotic plaques was achieved by cRGD conjugated liposomes (IL10-cRGD-Lip). RESULTS The IL10-cRGD-Lip of size 179.4 ± 10.91 nm having PDI 0.14 ± 0.04 with a surface charge of +18.34 ± 1.36 mV was prepared. The in-vitro analysis clearly suggests that IL10-cRGD-Lip sustains the release of IL10 and could significantly reduce ROS and NO. The immuno-staining results revealed that IL-1β and TNF-α were down-regulated after the treatment with IL10-cRGD-Lip in Lipopolysaccharide (LPS) stimulated RAW 264.7 cells. CONCLUSION the in-vitro results clearly suggest that anti-inflammatory cytokine IL10 could be used for the cure of inflammatory maladies including atherosclerosis.
Collapse
Affiliation(s)
- Jianchao Li
- Department of Cardiopulmonary Bypass, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuyan Ding
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoliang Qian
- Department of Cardiopulmonary Bypass, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Junjie Sun
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenwei Ge
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Leiyi Yang
- Department of Cardiopulmonary Bypass, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhaoyun Cheng
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
81
|
Bhatt P, Sharma M, Sharma S. Prediction and identification of T cell epitopes of COVID-19 with balanced cytokine response for the development of peptide based vaccines. In Silico Pharmacol 2021; 9:40. [PMID: 34221846 PMCID: PMC8237047 DOI: 10.1007/s40203-021-00098-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/05/2021] [Indexed: 12/27/2022] Open
Abstract
Recent outbreak of 2019 novel Corona virus poses serious challenge for the global health system. In lieu of paucity of experimental data, tools and the very basic understanding of host immune responses against SARS-CoV-2, well thought effective measures are needed to control COVID-19 pandemic. We have identified specific overlapping antigenic peptide epitopes (OAPE) within the 4 structural proteins of SARS-CoV-2 predictive of triggering robust CD4 and CD8 T cell responses in host using bio-informatics tools (NetMHC4.0, IEDB, and Vaxijen2.0). We speculate an early release of pro-inflammatory cytokines for protection and later release of anti-inflammatory cytokines for prevention of immunopathology in designing a vaccine for Covid-19. Therefore, the selected immunogenic OAPE were subjected to in silico tools (IL-6-Pred, IFNepitope and PIP-EL) for analyzing their pro-inflammatory response. The OAPEs found to be pro-inflammatory in nature were further subjected to prediction servers (IL-4-Pred, IL-10-Pred, Pre-AIP) to characterize them as inducers of anti-inflammatory response as well. We finally filtered out 12 OAPE which had affinity for both CD4 and CD8 T cells as well as were inducers of pro-inflammatory and anti-inflammatory cytokines. On confirmation of OAPE binding affinity for respective T cell specific MHC allele using docking studies (pepATTRACT, Hex8.0 and Discovery studio) they were found to be have more immunogenic potential than the 3 negative control peptides (NCPs) included in the study. Additionally, we constructed CTxB-adjuvanated multi-epitopic vaccine inclusive of the 12 OAPEs which was non-toxic, non-allergenic and capable of inducing both pro-inflammatory and anti-inflammatory cytokines. A successful in silico cloning and docking of modeled subunit vaccine construct with toll like receptor-2 (TLR-2) confirmed the high efficacy of our multi-epitopic vaccine which can through a balanced interplay of cytokines help in creating a steady-state immune equilibrium. In silico immune simulation studies with the vaccine using C-ImmSim server also showed higher percentage of T cells along with production of pro-inflammatory as well as some anti-inflammatory cytokines. Experimental validation of this prediction based study on Peripheral Blood Mononuclear Cells (PBMCs) of un-infected individuals, patients and recovered individuals will facilitate production of high priority effective SARS -CoV-2 vaccine candidate. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-021-00098-7.
Collapse
Affiliation(s)
- Parul Bhatt
- DS Kothari Central Facility for Interdisciplinary Research, Miranda House, University of Delhi, Delhi, 110007 India
- Department of Zoology, Miranda House, University of Delhi, Delhi, 110007 India
| | - Monika Sharma
- DS Kothari Central Facility for Interdisciplinary Research, Miranda House, University of Delhi, Delhi, 110007 India
- Department of Zoology, Miranda House, University of Delhi, Delhi, 110007 India
| | - Sadhna Sharma
- DS Kothari Central Facility for Interdisciplinary Research, Miranda House, University of Delhi, Delhi, 110007 India
- Department of Zoology, Miranda House, University of Delhi, Delhi, 110007 India
| |
Collapse
|
82
|
Zhu Z, Parikh P, Zhao H, Givens NT, Beck DB, Willson CM, Bai Q, Wakefield MR, Fang Y. Targeting immunometabolism of neoplasms by interleukins: A promising immunotherapeutic strategy for cancer treatment. Cancer Lett 2021; 518:94-101. [PMID: 34153401 DOI: 10.1016/j.canlet.2021.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/23/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022]
Abstract
In recent years, tumor metabolism has become a prevalent research topic for scientists and pharmaceutical companies. As research in the field has progressed, the metabolism-based therapy of tumors has ushered in new opportunities. Most tumors emerge and evolve under selective pressure from their microenvironment, which promotes the diversification of both neoplastic and non-neoplastic compartments of the tumor microenvironment (TME), and finally reaches a certain degree of intratumoral heterogeneity. As a result of the tumor intratumoral heterogeneity, tumor cells often possess a complex energy metabolism phenotype. During tumor progression, the metabolism for both tumor parenchyma and stroma is reprogrammed. The tumor stroma mainly consists of the extracellular matrix, fibroblasts, and immune cells. Interestingly, tumor-infiltrating immune cells utilize different metabolites based on their subtype and function, and these immunometabolic pathways can be modified in the TME. In particular, interleukins play a vital role in the activation and differentiation of immune cells and have exhibited multiple effects on tumor cell neoplasia, invasion, and metastasis. In this review, we summarize the common mechanisms of interleukins affecting the tumor and tumor-infiltrating immune cells metabolically and discuss how these mechanisms may lead to novel therapeutic opportunities. This review might contribute to the novel development of cancer immunotherapy.
Collapse
Affiliation(s)
- Ziwen Zhu
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA, USA; Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA.
| | - Pooja Parikh
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Hongyun Zhao
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nathan T Givens
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA, USA
| | - Damien B Beck
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA, USA
| | - Conner M Willson
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA, USA
| | - Qian Bai
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA, USA; Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA.
| |
Collapse
|
83
|
Saneei Totmaj A, Haghighat S, Jaberzadeh S, Navaei M, Vafa S, Janani L, Emamat H, Salehi Z, Izad M, Zarrati M. The Effects of Synbiotic Supplementation on Serum Anti-Inflammatory Factors in the Survivors of Breast Cancer with Lymphedema following a Low Calorie Diet: A Randomized, Double-Blind, Clinical Trial. Nutr Cancer 2021; 74:869-881. [PMID: 34085881 DOI: 10.1080/01635581.2021.1933096] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIM Breast cancer-related lymphedema (BCRL) is a treatment-related inflammatory complication in breast cancer survivors (BCSs). This study was aimed to evaluate the effect of synbiotic supplementation on serum concentrations of IL-10, TGF-β, VEGF, adiponectin, and edema volume among overweight or obese BCSs with lymphedema following a low-calorie diet (LCD). METHOD In a randomized double-blind, controlled clinical trial, 88 obese and overweight BCSs women were randomized to synbiotic supplement (n = 44) or placebo (n = 44) groups and both groups followed an LCD for 10 weeks. Pre- and post-intervention comparisons were made regarding the anti-inflammatory markers which included IL-10, TGF-β, VEGF, adiponectin, edema volume, and anthropometric measurements. Also, the same factors were analyzed to find inter-group disparities. RESULTS There were no significant differences among participants in the baseline, except for IL-10 and adiponectin. Post-intervention, no significant differences were observed regarding the anti-inflammatory markers, including IL-10, VEGF, adiponectin, and TGF-β between the groups. After 10 weeks of intervention edema volume significantly decreased in the synbiotic group; additionally, anthropometric measurements (body weight, BMI, body fat percent, and WC) decreased in both groups significantly (P < 0.001 and P < 0.005; respectively). CONCLUSION Synbiotic supplementation coupled with an LCD in a 10-week intervention had beneficial effects on increasing the serum TGF-β, IL-10, and adiponectin levels in women with BCRL. It also reduced arm lymphedema volume. Therefore, synbiotic supplementation can be effective in improving health status in BCRL patients.
Collapse
Affiliation(s)
- Ali Saneei Totmaj
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Shahpar Haghighat
- Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Shapour Jaberzadeh
- Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Australia
| | - Mehraban Navaei
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Saeideh Vafa
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Janani
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Hadi Emamat
- Student Research Committee, Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Immunology Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Izad
- Immunology Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Zarrati
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
84
|
Abstract
PURPOSE OF REVIEW The use of cytokines in harnessing the immune system to eradicate cancer has been an important treatment modality. However, the dose-limiting toxicities of these cytokines limited their usage in clinic. Here, we review the basic biology of cytokines involved in the treatment of melanoma and discuss their therapeutic applications. Moreover, we describe several innovative technological approaches that have been developed to improve the pharmacokinetics, safety, and efficacy of these cytokines. RECENT FINDINGS The safety and the anti-tumor activity of newly engineered cytokines including PEGylated IL-2 (NKTR-214), PEGylated IL-10 (AM0010), and IL-15 super agonist (ALT-803) have been evaluated in clinical trials with encouraging clinical activity and acceptable safety profile, both as single agents and in combination with immuno-oncology agents. A greater understanding of the mechanisms of action and effective dosing of these newly engineered cytokine together with determination of optimum combination therapy regimens may yield greater clinical benefits in the future.
Collapse
|
85
|
Parveen S, Siddharth S, Cheung LS, Kumar A, Shen J, Murphy JR, Sharma D, Bishai WR. Therapeutic targeting with DABIL-4 depletes myeloid suppressor cells in 4T1 triple-negative breast cancer model. Mol Oncol 2021; 15:1330-1344. [PMID: 33682324 PMCID: PMC8096791 DOI: 10.1002/1878-0261.12938] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/10/2021] [Accepted: 03/05/2021] [Indexed: 12/19/2022] Open
Abstract
In many solid tumors including triple-negative breast cancer (TNBC), upregulation of the interleukin-4 receptor (IL-4R) has been shown to promote cancer cell proliferation, apoptotic resistance, metastatic potential, and a Th2 response in the tumor microenvironment (TME). Since immunosuppressive cells in the TME and spleen including myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) also express the IL-4R, we hypothesized that selective depletion of IL-4R-bearing cells in TNBC would result in the direct killing of tumor cells and the depletion of immunosuppressive cells and lead to an enhanced antitumor response. To selectively target IL-4R+ cells, we employed DABIL-4, a fusion protein toxin consisting of the catalytic and translocation domains of diphtheria toxin fused to murine IL-4. As anticipated, DABIL-4 has potent cytotoxic activity against TNBC cells both in vitro and in vivo. We demonstrate in the murine 4T1 TNBC model that DABIL-4 significantly reduces tumor growth, splenomegaly, and lung metastases. Importantly, we also show that the administration of DABIL-4 results in the selective depletion of MDSCs, TAMs, and regulatory T cells in treated mice, with a concomitant increase in IFN-γ+ CD8 effector T cells in the TME. Since the 4T1 antitumor activity of DABIL-4 was largely diminished in IL-4R knockout mice, we postulate that DABIL-4 functions primarily as an immunotherapeutic by the depletion of MDSCs, TAMs, and regulatory T cells. NanoString analysis of control and treated tumors confirmed and extended these observations by showing a marked decline of mRNA transcripts that are associated with tumorigenesis and metastasis. In conclusion, we demonstrate that DABIL-4 targeting of both tumor and immunosuppressive host cells likely represents a novel and effective treatment strategy for 4T1 TNBC and warrants further study.
Collapse
Affiliation(s)
- Sadiya Parveen
- Department of MedicineDivision of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Sumit Siddharth
- Department of OncologySidney Kimmel Comprehensive Cancer Center at Johns HopkinsJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Laurene S. Cheung
- Department of MedicineDivision of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Alok Kumar
- Department of MedicineDivision of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Jessica Shen
- Department of MedicineDivision of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - John R. Murphy
- Department of MedicineDivision of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Dipali Sharma
- Department of OncologySidney Kimmel Comprehensive Cancer Center at Johns HopkinsJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - William R. Bishai
- Department of MedicineDivision of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
86
|
Yuba E, Budina E, Katsumata K, Ishihara A, Mansurov A, Alpar AT, Watkins EA, Hosseinchi P, Reda JW, Lauterbach AL, Nguyen M, Solanki A, Kageyama T, Swartz MA, Ishihara J, Hubbell JA. Suppression of Rheumatoid Arthritis by Enhanced Lymph Node Trafficking of Engineered Interleukin-10 in Murine Models. Arthritis Rheumatol 2021; 73:769-778. [PMID: 33169522 PMCID: PMC11095083 DOI: 10.1002/art.41585] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/13/2020] [Accepted: 11/05/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a major autoimmune disease that causes synovitis and joint damage. Although clinical trials have been performed using interleukin-10 (IL-10), an antiinflammatory cytokine, as a potential treatment of RA, the therapeutic effects of IL-10 have been limited, potentially due to insufficient residence in lymphoid organs, where antigen recognition primarily occurs. This study was undertaken to engineer an IL-10-serum albumin (SA) fusion protein and evaluate its effects in 2 murine models of RA. METHODS SA-fused IL-10 (SA-IL-10) was recombinantly expressed. Mice with collagen antibody-induced arthritis (n = 4-7 per group) or collagen-induced arthritis (n = 9-15 per group) were injected intravenously with wild-type IL-10 or SA-IL-10, and the retention of SA-IL-10 in the lymph nodes (LNs), immune cell composition in the paws, and therapeutic effect of SA-IL-10 on mice with arthritis were assessed. RESULTS SA fusion to IL-10 led to enhanced accumulation in the mouse LNs compared with unmodified IL-10. Intravenous SA-IL-10 treatment restored immune cell composition in the paws to a normal status, elevated the frequency of suppressive alternatively activated macrophages, reduced IL-17A levels in the paw-draining LN, and protected joint morphology. Intravenous SA-IL-10 treatment showed similar efficacy as treatment with an anti-tumor necrosis factor antibody. SA-IL-10 was equally effective when administered intravenously, locally, or subcutaneously, which is a benefit for clinical translation of this molecule. CONCLUSION SA fusion to IL-10 is a simple but effective engineering strategy for RA therapy and has potential for clinical translation.
Collapse
Affiliation(s)
- Eiji Yuba
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Department of Applied Chemistry, Osaka Prefecture University, Osaka 599-8531, Japan
| | - Erica Budina
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Kiyomitsu Katsumata
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ako Ishihara
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Aslan Mansurov
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Aaron T. Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Elyse A. Watkins
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Peyman Hosseinchi
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Joseph W. Reda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Abigail L. Lauterbach
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Mindy Nguyen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL 60637, USA
| | - Takahiro Kageyama
- Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Jun Ishihara
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
87
|
Witkowska-Piłaszewicz O, Pingwara R, Szczepaniak J, Winnicka A. The Effect of the Clenbuterol-β2-Adrenergic Receptor Agonist on the Peripheral Blood Mononuclear Cells Proliferation, Phenotype, Functions, and Reactive Oxygen Species Production in Race Horses In Vitro. Cells 2021; 10:cells10040936. [PMID: 33920705 PMCID: PMC8072563 DOI: 10.3390/cells10040936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
Clenbuterol, the β2-adrenoceptor agonist, is gaining growing popularity because of its effects on weight loss (i.e., chemical liposuction). It is also popular in bodybuilding and professional sports, due to its effects that are similar to anabolic steroids. However, it is prohibited by anti-doping control. On the other hand, it is suggested that clenbuterol can inhibit the inflammatory process. The cells from 14 untrained and 14 well-trained race horses were collected after acute exercise and cultured with clenbuterol. The expressions of CD4, CD8, FoxP3, CD14, MHCII, and CD5 in PBMC, and reactive oxygen species (ROS) production, as well as cell proliferation, were evaluated by flow cytometry. In addition, IL-1β, IL-4, IL-6, IL-10, IL-17, INF-γ and TNF-α concentrations were evaluated by ELISA. β2-adrenoceptor stimulation leads to enhanced anti-inflammatory properties in well-trained horses, as do low doses in untrained animals. In contrast, higher clenbuterol doses create a pro-inflammatory environment in inexperienced horses. In conclusion, β2-adrenoceptor stimulation leads to a biphasic response. In addition, the immune cells are more sensitive to drug abuse in inexperienced individuals under physical training.
Collapse
Affiliation(s)
- Olga Witkowska-Piłaszewicz
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Science—SGGW, 02-787 Warsaw, Poland;
- Correspondence:
| | - Rafał Pingwara
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, 02-787 Warsaw, Poland;
| | - Jarosław Szczepaniak
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences—SGGW, 02-787 Warsaw, Poland;
| | - Anna Winnicka
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Science—SGGW, 02-787 Warsaw, Poland;
| |
Collapse
|
88
|
Alberca GGF, Solis-Castro RL, Solis-Castro ME, Alberca RW. Coronavirus disease-2019 and the intestinal tract: An overview. World J Gastroenterol 2021; 27:1255-1266. [PMID: 33833480 PMCID: PMC8015300 DOI: 10.3748/wjg.v27.i13.1255] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/10/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection can progress to a severe respiratory and systemic disease named coronavirus disease-2019 (COVID-19). The most common symptoms are fever and respiratory discomfort. Nevertheless, gastrointestinal infections have been reported, with symptoms such as diarrhea, nausea, vomiting, abdominal pain, and lack of appetite. Importantly, SARS-CoV-2 can remain positive in fecal samples after nasopharyngeal clearance. After gastrointestinal SARS-CoV-2 infection and other viral gastrointestinal infections, some patients may develop alterations in the gastrointestinal microbiota. In addition, some COVID-19 patients may receive antibiotics, which may also disturb gastrointestinal homeostasis. In summary, the gastrointestinal system, gut microbiome, and gut-lung axis may represent an important role in the development, severity, and treatment of COVID-19. Therefore, in this review, we explore the current pieces of evidence of COVID-19 gastrointestinal manifestations, possible implications, and interventions.
Collapse
Affiliation(s)
- Gabriela Gama Freire Alberca
- Department of Microbiology, Institute of Biomedical Sciences-University of São Paulo, São Paulo 05508-000, Brazil
| | - Rosa Liliana Solis-Castro
- Departamento Académico de Biología Bioquímica, Facultad de Ciencias de la Salud, Universidad Nacional de Tumbes, Pampa Grande 24000, Tumbes, Peru
| | - Maria Edith Solis-Castro
- Departamento Académico de Medicina Humana, Facultad de Ciencias de la Salud, Universidad Nacional de Tumbes, Pampa Grande 24000, Tumbes, Peru
| | - Ricardo Wesley Alberca
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil
| |
Collapse
|
89
|
Wang H, Zhou Y, Sun Q, Zhou C, Hu S, Lenahan C, Xu W, Deng Y, Li G, Tao S. Update on Nanoparticle-Based Drug Delivery System for Anti-inflammatory Treatment. Front Bioeng Biotechnol 2021; 9:630352. [PMID: 33681167 PMCID: PMC7925417 DOI: 10.3389/fbioe.2021.630352] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/29/2021] [Indexed: 12/17/2022] Open
Abstract
Nanobiotechnology plays an important role in drug delivery, and various kinds of nanoparticles have demonstrated new properties, which may provide opportunities in clinical treatment. Nanoparticle-mediated drug delivery systems have been used in anti-inflammatory therapies. Diseases, such as inflammatory bowel disease, rheumatoid arthritis, and osteoarthritis have been widely impacted by the pathogenesis of inflammation. Efficient delivery of anti-inflammatory drugs can reduce medical dosage and improve therapeutic effect. In this review, we discuss nanoparticles with potential anti-inflammatory activity, and we present a future perspective regarding the application of nanomedicine in inflammatory diseases.
Collapse
Affiliation(s)
- Huailan Wang
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qunan Sun
- Department of Medical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chenghao Zhou
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiyao Hu
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Cameron Lenahan
- Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sifeng Tao
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
90
|
Karami E, Esfahrood ZR, Mansouri R, Haerian A, Abdian-Asl A. Effect of epigallocatechin-3-gallate on tumor necrosis factor-alpha production by human gingival fibroblasts stimulated with bacterial lipopolysaccharide: An in vitro study. J Indian Soc Periodontol 2021; 25:11-16. [PMID: 33642735 PMCID: PMC7904023 DOI: 10.4103/jisp.jisp_323_20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/24/2020] [Accepted: 10/11/2020] [Indexed: 11/26/2022] Open
Abstract
Background: Evidence shows that epigallocatechin-3-gallate (EGCG) in green tea has anti-inflammatory effects. Aim: This study assessed the effect of EGCG on the production of tumor necrosis factor-alpha (TNF-α) as an inflammatory cytokine in periodontitis, which produced by human gingival fibroblasts (HGFs) stimulated with lipopolysaccharide (LPS) of Porphyromonas gingivalis. Materials and Methods: In this study, HGFs were cultured and subjected to LPS and EGCG. Cell viability of different concentrations of EGCG (10, 25, 50, 75, and 100 μM) and LPS (1, 10, 20, and 50 μg/mL) was assessed using methyl-thiazole-tetrazolium (MTT) assay. Then, the best concentrations of EGCG and P. gingivalis LPS were used simultaneously and separately to assess the production of TNF-α by HGFs using the enzyme-linked immunosorbent assay (ELISA). Assessments were done at 1, 3, and 5 days. Data were read using the ELISA reader and analyzed by the SPSS through two-way ANOVA. Results: LPS at 1, 10, and 20 and EGCG at 10.25 and 50 μM showed the least cytotoxicity in MTT assay. ELISA showed EGCG alone decreased the production of TNF-α in all days, except 10 μM on day 1. 1, 10, and 20 μg/mL LPS increased the output of TNF-α on days 1 and 3 while reducing it on day 5. The combination of EGCG and LPS showed a decrease of TNF-α in all days except on day 5 that revealed an increase in the production of TNF-α at 25 and 50 μM EGCG. Conclusion: In the combination use of EGCG and LPS, EGCG shows anti-inflammatory effects by decreasing the production of TNF-α by HGFs stimulated with P. gingivalis.
Collapse
Affiliation(s)
- Elahe Karami
- Department of Periodontics, School of Dentistry, Shahid Sadughi University of Medical Science, Yazd, Iran
| | - Zeinab Rezaei Esfahrood
- Department of Periodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Mansouri
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Blood and oncology research center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ahmad Haerian
- Department of Periodontics, School of Dentistry, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Amir Abdian-Asl
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
91
|
Gunasinghe SD, Peres NG, Goyette J, Gaus K. Biomechanics of T Cell Dysfunctions in Chronic Diseases. Front Immunol 2021; 12:600829. [PMID: 33717081 PMCID: PMC7948521 DOI: 10.3389/fimmu.2021.600829] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Understanding the mechanisms behind T cell dysfunctions during chronic diseases is critical in developing effective immunotherapies. As demonstrated by several animal models and human studies, T cell dysfunctions are induced during chronic diseases, spanning from infections to cancer. Although factors governing the onset and the extent of the functional impairment of T cells can differ during infections and cancer, most dysfunctional phenotypes share common phenotypic traits in their immune receptor and biophysical landscape. Through the latest developments in biophysical techniques applied to explore cell membrane and receptor-ligand dynamics, we are able to dissect and gain further insights into the driving mechanisms behind T cell dysfunctions. These insights may prove useful in developing immunotherapies aimed at reinvigorating our immune system to fight off infections and malignancies more effectively. The recent success with checkpoint inhibitors in treating cancer opens new avenues to develop more effective, targeted immunotherapies. Here, we highlight the studies focused on the transformation of the biophysical landscape during infections and cancer, and how T cell biomechanics shaped the immunopathology associated with chronic diseases.
Collapse
Affiliation(s)
- Sachith D Gunasinghe
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Newton G Peres
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Jesse Goyette
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
92
|
Cappelli K, Sabino M, Trabalza-Marinucci M, Acuti G, Capomaccio S, Menghini L, Verini-Supplizi A. Differential Effects of Dietary Oregano Essential Oil on the Inflammation Related Gene Expression in Peripheral Blood Mononuclear Cells From Outdoor and Indoor Reared Pigs. Front Vet Sci 2021; 8:602811. [PMID: 33718464 PMCID: PMC7946822 DOI: 10.3389/fvets.2021.602811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Intensive farming systems represent a stressful environment for pigs and negatively influence neuroendocrine functions, behavior, and performance. Outdoor farming is an alternative option, which is thought to imply several beneficial effects for the animal. Dietary essential oils are known to be an innovative strategy to improve pig health and performance, and oregano essential oil (ORE) possesses beneficial effects due to its antimicrobial, anti-fungal, and antioxidant properties. We tested the effect of dietary ORE on peripheral blood mononuclear cells (PBMCs) in 36 growing pigs, either reared under indoor or outdoor conditions. Quantitative real-time PCR (RT-qPCR) assay was used to evaluate the effect of diet (control vs. ORE) and the time of sampling (T1-120 days vs. T2-190 days) on the expression of inflammatory and immune-related genes (TNF, IL1β, IL8, IL18, IL10, IL1RN, STAT3, HSP90, ICAM-1, and NFKB1). Under outdoor condition, the majority of transcripts were upregulated (p < 0.05), assuming a general inflammatory status (TNF, HSP90, NFKB1, IL1β, and STAT3). However, an interaction between diet and the farming system was observed: HSP90, NFKB1, and STAT3 were downregulated (p < 0.05) in the outdoor reared pigs when fed the ORE diet. Our study showed that bioactive compounds of ORE exert their activity, especially when the animals are exposed to stressful stimuli. Dietary ORE can be an acceptable strategy to help pigs tolerate the stress related to the harsh, outdoor, rearing conditions.
Collapse
Affiliation(s)
- Katia Cappelli
- Dipartimento di Medicina Veterinaria, University of Perugia, Perugia, Italy
| | - Marcella Sabino
- Dipartimento di Medicina Veterinaria, University of Perugia, Perugia, Italy
| | | | - Gabriele Acuti
- Dipartimento di Medicina Veterinaria, University of Perugia, Perugia, Italy
| | - Stefano Capomaccio
- Dipartimento di Medicina Veterinaria, University of Perugia, Perugia, Italy
| | - Luigi Menghini
- Dipartimento di Farmacia, University of Chieti, Chieti, Italy
| | | |
Collapse
|
93
|
Bergens O, Nilsson A, Papaioannou KG, Kadi F. Sedentary Patterns and Systemic Inflammation: Sex-Specific Links in Older Adults. Front Physiol 2021; 12:625950. [PMID: 33613317 PMCID: PMC7892961 DOI: 10.3389/fphys.2021.625950] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/15/2021] [Indexed: 11/13/2022] Open
Abstract
The study aimed to examine sex-specific associations between objectively measured sedentary patterns and pro- and anti-inflammatory biomarkers in older adults when considering the moderating impact of physical activity (PA). Accelerometer-based monitoring of sedentary patterns and PA was conducted in a population of older men (n = 83; age: 67.4 ± 1.5; height: 178.7 ± 6.6 cm; weight: 80.9 ± 10.6 kg) and women (n = 146; age: 67.4 ± 1.6; height: 164.2 ± 6.1 cm; weight: 64.6 ± 10.1 kg) aged 65-70. Blood samples were collected for the assessment of the inflammatory biomarkers C-reactive protein (CRP), fibrinogen, interleukin-6 (IL-6), IL-10, IL-18, and monocyte chemoattractant protein-1 (MCP-1). Data were analyzed using multiple linear regression models. Total and bouts of ≥10 min of sedentary time were inversely associated with the anti-inflammatory marker IL-10 in older men (accumulated sedentary time: β = -0.116; bouts: β = -0.099; all p < 0.05). Associations were independent of moderate-to-vigorous physical activity (MVPA) and total PA volume. In women, total and bouts of ≥10 min of sedentary time were detrimentally associated with the pro-inflammatory marker fibrinogen (accumulated sedentary time: β = -0.130; bouts: β = -0.085; all p < 0.05). Associations remained between accumulated sedentary time and fibrinogen when adjusting for MVPA and total PA volume. This study highlights sex-specific routes by which sedentary patterns impact on pro- and anti-inflammatory biomarkers in older adults. The findings support efforts to promote accumulation of time spent in PA at the expense of time in sedentary pursuits on low-grade inflammation in older men and women.
Collapse
Affiliation(s)
- Oscar Bergens
- School of Health Sciences, Örebro University, Örebro, Sweden
| | - Andreas Nilsson
- School of Health Sciences, Örebro University, Örebro, Sweden
| | | | - Fawzi Kadi
- School of Health Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
94
|
Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2021; 1:493-506. [PMID: 22754768 PMCID: PMC3382908 DOI: 10.4161/onci.20459] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
During the last two decades, a number of approaches for the activation of the immune system against cancer has been developed. These include highly specific interventions, such as monoclonal antibodies, vaccines and cell-based therapies, as well as relatively unselective strategies, such as the systemic administration of adjuvants and immunomodulatory cytokines. Cytokines constitute a huge group of proteins that, taken together, regulate not only virtually all the aspects of innate and cognate immunity, but also several other cellular and organismal functions. Cytokines operate via specific transmembrane receptors that are expressed on the plasma membrane of target cells and, depending on multiple variables, can engage autocrine, paracrine or endocrine signaling pathways. The most appropriate term for defining the cytokine network is “pleiotropic”: cytokines are produced by - and operate on - multiple, often overlapping, cell types, triggering context-depend biological outcomes as diverse as cell proliferation, chemotaxis, differentiation, inflammation, elimination of pathogens and cell death. Moreover, cytokines often induce the release of additional cytokines, thereby engaging self-amplificatory or self-inhibitory signaling cascades. In this Trial Watch, we will summarize the biological properties of cytokines and discuss the progress of ongoing clinical studies evaluating their safety and efficacy as immunomodulatory agents against cancer.
Collapse
Affiliation(s)
- Erika Vacchelli
- INSERM; U848; Villejuif, France ; Université Paris-Sud/Paris XI; Paris, France
| | | | | | | | | | | | | |
Collapse
|
95
|
Pham PN, Huličiak M, Biedermannová L, Černý J, Charnavets T, Fuertes G, Herynek Š, Kolářová L, Kolenko P, Pavlíček J, Zahradník J, Mikulecky P, Schneider B. Protein Binder (ProBi) as a New Class of Structurally Robust Non-Antibody Protein Scaffold for Directed Evolution. Viruses 2021; 13:v13020190. [PMID: 33514045 PMCID: PMC7911045 DOI: 10.3390/v13020190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/15/2021] [Accepted: 01/23/2021] [Indexed: 12/13/2022] Open
Abstract
Engineered small non-antibody protein scaffolds are a promising alternative to antibodies and are especially attractive for use in protein therapeutics and diagnostics. The advantages include smaller size and a more robust, single-domain structural framework with a defined binding surface amenable to mutation. This calls for a more systematic approach in designing new scaffolds suitable for use in one or more methods of directed evolution. We hereby describe a process based on an analysis of protein structures from the Protein Data Bank and their experimental examination. The candidate protein scaffolds were subjected to a thorough screening including computational evaluation of the mutability, and experimental determination of their expression yield in E. coli, solubility, and thermostability. In the next step, we examined several variants of the candidate scaffolds including their wild types and alanine mutants. We proved the applicability of this systematic procedure by selecting a monomeric single-domain human protein with a fold different from previously known scaffolds. The newly developed scaffold, called ProBi (Protein Binder), contains two independently mutable surface patches. We demonstrated its functionality by training it as a binder against human interleukin-10, a medically important cytokine. The procedure yielded scaffold-related variants with nanomolar affinity.
Collapse
|
96
|
Han QQ, Yin M, Wang ZY, Liu H, Ao JP, Wang YX. Cynandione A Alleviates Neuropathic Pain Through α7-nAChR-Dependent IL-10/β-Endorphin Signaling Complexes. Front Pharmacol 2021; 11:614450. [PMID: 33584292 PMCID: PMC7873367 DOI: 10.3389/fphar.2020.614450] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022] Open
Abstract
Cynandione A, an acetophenone isolated from Cynanchum Wilfordii Radix, exhibits antineuropathic pain effect. This study further explored the target molecule and signaling mechanisms underlying cynandione-A-induced antineuropathic pain. Intrathecal injection of cynandione A significantly attenuated mechanical allodynia in neuropathic rats and substantially increased spinal expression of IL-10 and β-endorphin but not dynorphin A. Cynandione A treatment also enhanced expression of IL-10 and β-endorphin but not α7 nicotinic acetylcholine receptors (nAChRs) in cultured microglia. The IL-10 antibody attenuated cynandione-A-induced spinal or microglial gene expression of β-endorphin and mechanical allodynia, whereas the β-endorphin antiserum blocked cynandione-A-induced mechanical antiallodynia but not spinal or microglial IL-10 gene expression. The α7 nAChR antagonist methyllycaconitine significantly reduced cynandione-A-induced mechanical antiallodynia and spinal or microglial expression of IL-10 and β-endorphin. Furthermore, cynandione A stimulated microglial phosphorylation of PKA, p38, and CREB in an α7-nAChR-dependent manner, and treatment with their inhibitors attenuated cynandione-A-induced mechanical antiallodynia and spinal or microglial expression of IL-10 and β-endorphin. In addition, cynandione A stimulated spinal phosphorylation of the transcription factor STAT3, which was inhibited by methyllycaconitine, the PKA activation inhibitor or IL-10 antibody. The STAT3 inhibitor NSC74859 also abolished cynandione-A-induced mechanical antiallodynia and spinal expression of β-endorphin. These findings suggest that cynandione A suppresses neuropathic pain through α7-nAChR-dependent IL-10/β-endorphin signaling pathway in spinal microglia.
Collapse
Affiliation(s)
- Qiao-Qiao Han
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai, China
| | - Min Yin
- Jiangsu Key Laboratory for the Research and Utilization of Plants Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
| | - Zi-Ying Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai, China
| | - Hao Liu
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai, China
| | - Jun-Ping Ao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yong-Xiang Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai, China
| |
Collapse
|
97
|
Wang K, Jiao Z, Chen H, Liu X, Lu J, Liu X, Li J, Wang L. The association between rs1800872 polymorphism in interleukin-10 and risk of cervical cancer: A meta-analysis. Medicine (Baltimore) 2021; 100:e23892. [PMID: 33545957 PMCID: PMC7837934 DOI: 10.1097/md.0000000000023892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In recent years, several reports have tried to prove this connection between rs1800872 polymorphism in interleukin-10 and cervical cancer among different populations, but the results are debatable. Thus, we collected all the published literature and conducted an integrated meta-analysis, which provided better evidence-based medicine for the relationship between rs1800872 polymorphism in interleukin-10 and risk of cervical cancer. METHODS We systematically performed our search on PubMed, EMBASE, Web of Science, WanFang database, and CNKI for all papers related to this research, published up to August 1, 2020. Summary odds ratios (OR) with 95% confidence interval (95% CI) were calculated in allelic, homozygous, heterozygous, dominant, and recessive model to appraise the association. RESULTS The meta-analysis included 8 studies containing 1393 cervical cancer cases and 1307 controls. The aggregate data under heterozygous model and dominant inheritance model (OR = 0.66, 95% CI: 0.55--0.80) indicated a significant association between rs1800872 and the low risk of cervical cancer in the entire population. And the aggregated data under the dominant inheritance model shows that rs1800872 is significantly associated with the reduction in the risk of cervical tumors in the entire population. CONCLUSION Our conclusion is that the AC/AA + AC variant of Rs1800872 indicates a protective effect in the development of cervical cancer.
Collapse
|
98
|
Barroso FAL, de Jesus LCL, de Castro CP, Batista VL, Ferreira Ê, Fernandes RS, de Barros ALB, Leclerq SY, Azevedo V, Mancha-Agresti P, Drumond MM. Intake of Lactobacillus delbrueckii (pExu: hsp65) Prevents the Inflammation and the Disorganization of the Intestinal Mucosa in a Mouse Model of Mucositis. Microorganisms 2021; 9:microorganisms9010107. [PMID: 33466324 PMCID: PMC7824804 DOI: 10.3390/microorganisms9010107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
5-Fluorouracil (5-FU) is an antineoplastic drug that causes, as a side effect, intestinal mucositis, acute inflammation in the small bowel. The Heat Shock Protein (Hsp) are highly expressed in inflammatory conditions, developing an important role in immune modulation. Thus, they are potential candidates for the treatment of inflammatory diseases. In the mucositis mouse model, the present study aimed to evaluate the beneficial effect of oral administration of milk fermented by Lactobacillus delbrueckii CIDCA 133 (pExu:hsp65), a recombinant strain. This approach showed increased levels of sIgA in the intestinal fluid, reducing inflammatory infiltrate and intestinal permeability. Additionally, the histological score was improved. Protection was associated with a reduction in the gene expression of pro-inflammatory cytokines such as Tnf, Il6, Il12, and Il1b, and an increase in Il10, Muc2, and claudin 1 (Cldn1) and 2 (Cldn2) gene expression in ileum tissue. These findings are corroborated with the increased number of goblet cells, the electronic microscopy images, and the reduction of intestinal permeability. The administration of milk fermented by this recombinant probiotic strain was also able to reverse the high levels of gene expression of Tlrs caused by the 5-FU. Thus, the rCIDCA 133:Hsp65 strain was revealed to be a promising preventive strategy for small bowel inflammation.
Collapse
Affiliation(s)
- Fernanda Alvarenga Lima Barroso
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Luís Cláudio Lima de Jesus
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Camila Prosperi de Castro
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Viviane Lima Batista
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Ênio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Renata Salgado Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus da UFMG, Universidade Federal de Minas Gerais, Cidade Universitária, Belo Horizonte 31270-901, Brazil; (R.S.F.); (A.L.B.d.B.)
| | - André Luís Branco de Barros
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus da UFMG, Universidade Federal de Minas Gerais, Cidade Universitária, Belo Horizonte 31270-901, Brazil; (R.S.F.); (A.L.B.d.B.)
| | - Sophie Yvette Leclerq
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel Dias (FUNED), Belo Horizonte 30510-010, Brazil;
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Pamela Mancha-Agresti
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
- Faculdade de Minas-Faminas-BH, Medicina, Belo Horizonte 31744-007, Brazil
- Correspondence: (P.M.-A.); (M.M.D.); Tel.: +55-31-99817-5004 (P.M.-A.); +55-31-99222-2761 (M.M.D.)
| | - Mariana Martins Drumond
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Departamento de Ciências Biológicas, Belo Horizonte 31421-169, Brazil
- Correspondence: (P.M.-A.); (M.M.D.); Tel.: +55-31-99817-5004 (P.M.-A.); +55-31-99222-2761 (M.M.D.)
| |
Collapse
|
99
|
Martinez JO, Evangelopoulos M, Brozovich AA, Bauza G, Molinaro R, Corbo C, Liu X, Taraballi F, Tasciotti E. Mesenchymal Stromal Cell‐Mediated Treatment of Local and Systemic Inflammation through the Triggering of an Anti‐Inflammatory Response. ADVANCED FUNCTIONAL MATERIALS 2021; 31. [DOI: 10.1002/adfm.202002997] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Indexed: 01/05/2025]
Abstract
AbstractThe emergence of cell‐based therapeutics, specifically the use of mesenchymal stromal/stem cells (MSCs), stands to significantly affect the future of targeted drug delivery technologies. MSCs represent a unique cell type, offering more than only regenerative potential but also site‐specific inflammatory targeting and tissue infiltration. In this study, a versatile multicomponent delivery platform, combining MSC tropism with multistage nanovector (MSV)‐mediated payload delivery, is debuted. It is demonstrated that the incorporation of drug‐loaded MSVs bestows MSCs with the ability to transport anti‐inflammatory payloads, achieving a fivefold increase in payload release without negatively impacting cellular functions, viability, extravasation, and inflammatory homing. When incorporated within MSCs, MSVs avoid rapid sequestration by filtering organs and conserve a 15‐fold increase in local inflammatory targeting compared to healthy ears. Furthermore, this MSC‐mediated MSV platform (M&Ms) rapidly triggers a 4.5‐fold reduction of local inflammation compared to free drug and extends survival to 100% of treated mice in a lethal model of systemic inflammation.
Collapse
Affiliation(s)
- Jonathan O. Martinez
- Center for Musculoskeletal Regeneration Houston Methodist Research Institute 6670 Bertner Ave Houston Houston TX 77030 USA
| | - Michael Evangelopoulos
- Center for Musculoskeletal Regeneration Houston Methodist Research Institute 6670 Bertner Ave Houston Houston TX 77030 USA
| | - Ava A. Brozovich
- Center for Musculoskeletal Regeneration Houston Methodist Research Institute 6670 Bertner Ave Houston Houston TX 77030 USA
- Texas A&M College of Medicine 8447 Bryan Rd, Bryan Houston TX 77807 USA
- Orthopedics and Sports Medicine Houston Methodist Hospital 6565 Fannin Street Houston Houston TX 77030 USA
| | - Guillermo Bauza
- Center for Musculoskeletal Regeneration Houston Methodist Research Institute 6670 Bertner Ave Houston Houston TX 77030 USA
- Center for NanoHealth Swansea University Medical School Swansea University Bay, Singleton Park Swansea Wales SA2 8PP UK
| | - Roberto Molinaro
- Center for Musculoskeletal Regeneration Houston Methodist Research Institute 6670 Bertner Ave Houston Houston TX 77030 USA
| | - Claudia Corbo
- Center for Musculoskeletal Regeneration Houston Methodist Research Institute 6670 Bertner Ave Houston Houston TX 77030 USA
- School of Medicine and Surgery Nanomedicine Center NANOMIB University of Milano‐Bicocca Vedano al Lambro MB 20854 Italy
| | - Xuewu Liu
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration Houston Methodist Research Institute 6670 Bertner Ave Houston Houston TX 77030 USA
- Orthopedics and Sports Medicine Houston Methodist Hospital 6565 Fannin Street Houston Houston TX 77030 USA
| | - Ennio Tasciotti
- Center for Musculoskeletal Regeneration Houston Methodist Research Institute 6670 Bertner Ave Houston Houston TX 77030 USA
- Texas A&M College of Medicine 8447 Bryan Rd, Bryan Houston TX 77807 USA
- Orthopedics and Sports Medicine Houston Methodist Hospital 6565 Fannin Street Houston Houston TX 77030 USA
| |
Collapse
|
100
|
Hwang JT, Yu JW, Nam HJ, Song SK, Sung WY, Kim, Y, Cho JH. Suppressive Effects of a Truncated Inhibitor K562 Protein-Derived Peptide on Two Proinflammatory Cytokines, IL-17 and TNF-α. J Microbiol Biotechnol 2020; 30:1810-1818. [PMID: 32958733 PMCID: PMC9728226 DOI: 10.4014/jmb.2004.04062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Inhibitor K562 (IK) protein was first isolated from the culture medium of K562 cells, a leukemia cell line, and is an inhibitory regulator of interferon-γ-induced major histocompatibility complex class II expression. Recently, exogenous truncated IK (tIK) protein showed potential as a therapeutic agent for inflammation-related diseases. In this study, we designed a novel putative anti-inflammatory peptide derived from tIK protein based on homology modeling of the human interleukin-10 (hIL-10) structure, and investigated whether the peptide exerted inhibitory effects against proinflammatory cytokines such as IL-17 and tumor necrosis factor-α (TNF-α). The peptide contains key residues involved in binding hIL-10 to the IL-10 receptor, and exerted strong inhibitory effects on IL- 17 (43.8%) and TNF-α (50.7%). In addition, we used circular dichroism spectroscopy to confirm that the peptide is usually present in a random coil configuration in aqueous solution. In terms of toxicity, the peptide was found to be biologically safe. The mechanisms by which the short peptide derived from human tIK protein exerts inhibitory effects against IL-17 and TNF-α should be explored further. We also evaluated the feasibility of using this novel peptide in skincare products.
Collapse
Affiliation(s)
- Jong Tae Hwang
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Ji Won Yu
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea,Department of Biology, Kongju National University, Kongju 3588, Republic of Korea
| | - Hee Jin Nam
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Sun Kwang Song
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Woo Yong Sung
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Yongae Kim,
- Department of Chemistry, Hankuk University of Foreign Studies, Yongin 1705, Republic of Korea
| | - Jang-Hee Cho
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea,Corresponding author Phone: +82-31-695-7959 Fax: +82-31-695-7986 E-mail:
| |
Collapse
|