151
|
Rong YD, Bian AL, Hu HY, Ma Y, Zhou XZ. Study on relationship between elderly sarcopenia and inflammatory cytokine IL-6, anti-inflammatory cytokine IL-10. BMC Geriatr 2018; 18:308. [PMID: 30541467 PMCID: PMC6292155 DOI: 10.1186/s12877-018-1007-9] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 12/05/2018] [Indexed: 12/16/2022] Open
Abstract
Background and objectives The pathophysiological mechanism of sarcopenia in the elderly has not yet been fully understood. Here, we aim to explore the relationship between sarcopenia and the inflammatory cytokine interleukin-6 (IL-6), and the anti-inflammatory cytokine interleukin-10 (IL-10) in an elderly population. Methods Our study comprised 118 males and 46 females aged between 61 and 90 who had received a general medical examination in Tianjin First Central Hospital. Subjects were divided into a sarcopenia group and a non-sarcopenia group, defined according to the criteria of the Asian Working Group for Sarcopenia (AWGS). We compared body composition, handgrip strength (HS), gait speed (GS), biochemical indexes, levels of IL-6 and IL-10, living habits, and disease status between these groups. Results Non-sarcopenia subjects undertook more regular physical exercise than sarcopenia patients. Sarcopenia subjects had higher nutrition risk but lower body mass index (BMI), serum albumin (ALB), triglyceride (TG), and creatinine (Cr) levels compared to non-sarcopenia subjects. Sarcopenia subjects were older and had higher visceral fat tissue (VFA) than non-sarcopenia subjects (P < 0.05), along with higher IL-6 and IL-10 levels. Furthermore, IL-6/IL-10 ratios were higher in subjects with sarcopenia (P < 0.05). Age, BMI, levels of physical activity, nutritional risk, VFA, IL-6, IL-10, IL-6/IL-10 ratio were independently associated with the presence of sarcopenia in univariate regression analyses. Following adjustment for confounding factors, the presence of sarcopenia was positively correlated with IL-6, IL-10, IL-6/IL-10 ratio and inversely correlated with BMI. Age is associated with increased presence of sarcopenia. Conclusions The levels of inflammation cytokine IL-6, anti-inflammatory IL-10 and IL-6/IL-10 ratio were increased in elderly sarcopenia subjects. Sarcopenia was associated with increased levels of inflammatory cytokine IL-6, anti-inflammatory cytokine IL-10, and IL-6/IL-10 ratios.
Collapse
Affiliation(s)
- Yu-Dong Rong
- Health Management Centre, Tianjin First Central Hospital, No. 24 of Fukang Road, Nankai District, Tianjin, 300192, China.
| | - Ai-Lin Bian
- Department of Geratology Tianjin First Central Hospital, Tianjin, China
| | - Hui-Ying Hu
- Department of Geratology Tianjin First Central Hospital, Tianjin, China
| | - Yue Ma
- Department of Geratology Yanda International Hospital, Hebei, China
| | - Xin-Zi Zhou
- Department of Geratology Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
152
|
Vonberg AD, Acevedo-Calado M, Cox AR, Pietropaolo SL, Gianani R, Lundy SK, Pietropaolo M. CD19+IgM+ cells demonstrate enhanced therapeutic efficacy in type 1 diabetes mellitus. JCI Insight 2018; 3:99860. [PMID: 30518692 DOI: 10.1172/jci.insight.99860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
We describe a protective effect on autoimmune diabetes and reduced destructive insulitis in NOD.scid recipients following splenocyte injections from diabetic NOD donors and sorted CD19+ cells compared with NOD.scid recipients receiving splenocytes alone. This protective effect was age specific (only CD19+ cells from young NOD donors exerted this effect; P < 0.001). We found that the CD19+IgM+ cell is the primary subpopulation of B cells that delayed transfer of diabetes mediated by diabetogenic T cells from NOD mice (P = 0.002). Removal of IgM+ cells from the CD19+ pool did not result in protection. Notably, protection conferred by CD19+IgM+ cotransfers were not dependent on the presence of Tregs, as their depletion did not affect their ability to delay onset of diabetes. Blockade of IL-10 with neutralizing antibodies at the time of CD19+ cell cotransfers also abrogated the therapeutic effect, suggesting that IL-10 secretion was an important component of protection. These results were strengthened by ex vivo incubation of CD19+ cells with IL-5, resulting in enhanced proliferation and IL-10 production and equivalently delayed diabetes progression (P = 0.0005). The potential to expand CD19+IgM+ cells, especially in response to IL-5 stimulation or by pharmacologic agents, may be a new therapeutic option for type 1 diabetes.
Collapse
Affiliation(s)
- Andrew D Vonberg
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Maria Acevedo-Calado
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Aaron R Cox
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Susan L Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Roberto Gianani
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Massimo Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| |
Collapse
|
153
|
Kina S, Kinjo T, Liang F, Nakasone T, Yamamoto H, Arasaki A. Targeting EphA4 abrogates intrinsic resistance to chemotherapy in well-differentiated cervical cancer cell line. Eur J Pharmacol 2018; 840:70-78. [DOI: 10.1016/j.ejphar.2018.09.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/12/2018] [Accepted: 09/26/2018] [Indexed: 01/12/2023]
|
154
|
Barroso-Sousa R, Ott PA. Transformation of Old Concepts for a New Era of Cancer Immunotherapy: Cytokine Therapy and Cancer Vaccines as Combination Partners of PD1/PD-L1 Inhibitors. Curr Oncol Rep 2018; 21:1. [PMID: 30498900 DOI: 10.1007/s11912-018-0738-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICI) are only effective in a subset of patients. Here, we will review the rationale and data supporting the combination of PD-1 pathway inhibition with recombinant cytokines and neoantigen-based cancer vaccines that can potentially increase the number of patients who will benefit from immunotherapy. RECENT FINDINGS The safety and tolerability of new interleukin(IL)-2 formulations, IL-15 super agonist, and PEGylated IL-10 have been evaluated in early phase clinical trials with promising efficacy data, both as monotherapy and in combination with ICI. Larger studies focusing on the efficacy of these combinations are ongoing. Personalized neoantigen-based cancer vaccines, enabled by improvements in sequencing computational capabilities, have been proven to be feasible, safe, and able to trigger a consistent vaccine-specific immune response in cancer patients. New pharmacologically modified recombinant cytokines and personalized neoantigen-based vaccines may turn these approaches into powerful tools for effective combination immunotherapy.
Collapse
Affiliation(s)
- Romualdo Barroso-Sousa
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana 2127, Boston, MA, 02215, USA
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana 2127, Boston, MA, 02215, USA.
| |
Collapse
|
155
|
Wąsik M, Nazimek K, Bryniarski K. Regulatory B cell phenotype and mechanism of action: the impact of stimulating conditions. Microbiol Immunol 2018; 62:485-496. [PMID: 29998521 DOI: 10.1111/1348-0421.12636] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/29/2018] [Accepted: 07/09/2018] [Indexed: 12/14/2022]
Abstract
A diverse population of regulatory B (Breg) cells reportedly exhibits significant immunomodulatory effects in various models of inflammatory responses and infectious diseases caused by bacteria, viruses or parasites. Breg cells contribute to maintenance of homeostasis via IL-10 production and multiple IL-10-independent mechanisms. The current review describes various phenotypic and functional subsets of Breg cells in autoimmune and infectious diseases and discusses the impacts of experimental conditions that have been found to drive Breg cell differentiation.
Collapse
Affiliation(s)
- Magdalena Wąsik
- Department of Immunology, Jagiellonian University College of Medicine, 18 Czysta St., 31-121 Krakow, Poland
| | - Katarzyna Nazimek
- Department of Immunology, Jagiellonian University College of Medicine, 18 Czysta St., 31-121 Krakow, Poland
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University College of Medicine, 18 Czysta St., 31-121 Krakow, Poland
| |
Collapse
|
156
|
Chebotareva N, Bobkova I, Lysenko L, Neprinzeva N, Vinogradov A, Moiseev S. Heat shock protein 70 and anti-heat shock protein 70 antibodies in patients with chronic glomerulonephritis. Cell Stress Chaperones 2018; 23:1229-1235. [PMID: 30062391 PMCID: PMC6237676 DOI: 10.1007/s12192-018-0928-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 10/28/2022] Open
Abstract
We evaluated the heat shock system 70 (HSP70) in patients with chronic glomerulonephritis (CGN). Seventy-six patients with CGN patients were included in our study. Ten patients with mild proteinuria (median 0.48 [0.16-0.78] g/24 h) and ten healthy subjects served as positive and negative controls, respectively. Urinary levels of HSP70, interleukin-10, and serum levels of anti-HSP70 were measured by ELISA. The immunohistochemical peroxidase method was used to study the expression of HSP70 and Foxp3+ in kidney biopsies. TregFoxP3+ cells in the interstitium were determined morphometrically. Median urinary HSP70 levels in patients with nephrotic syndrome (NS) [6.57 (4.49-8.33) pg/mg] and subnephrotic range proteinuria [5.7 (4.12-6.9) pg/mg] were higher (p < 0.05) than in positive [3.7 (2.5-4.82) pg/mg] and negative [3.78 (2.89-4.84) pg/mg] controls. HSP70 expression index in tubular cells positively correlated with urinary HSP70 (Rs = 0.948, р < 0.05) and proteinuria (Rs = 0.362, p < 0.05). The number of TregFoxp3+ cells in the kidney interstitium and interleukin-10 excretion were lower in patients with NS. Anti-HSP70 antibody serum levels in patients with NS [21.1 (17.47-29.72) pg/ml] and subnephrotic range proteinuria [24.9 (18.86-30.92) pg/ml] were significantly higher than in positive [17.8 (12.95-23.03) pg/ml] and negative [18.9 (13.5-23.9) pg/ml] controls. In patients with CGN, increasing proteinuria was associated with higher HSP70 renal tissue and urinary levels. However, activation of HSP70 in patients with nephrotic syndrome did not lead to an increase in tissue levels of TregFoxp3+ cells or to the release of IL-10.
Collapse
Affiliation(s)
- Natalia Chebotareva
- Tareev Clinic of Internal Diseases, Sechenov First Moscow State Medical University, Rossolimo 11/5, Moscow, Russia, 119435.
| | - Irina Bobkova
- Tareev Clinic of Internal Diseases, Sechenov First Moscow State Medical University, Rossolimo 11/5, Moscow, Russia, 119435
| | - Lidia Lysenko
- Tareev Clinic of Internal Diseases, Sechenov First Moscow State Medical University, Rossolimo 11/5, Moscow, Russia, 119435
| | - Natalia Neprinzeva
- Tareev Clinic of Internal Diseases, Sechenov First Moscow State Medical University, Rossolimo 11/5, Moscow, Russia, 119435
| | - Anatoly Vinogradov
- Tareev Clinic of Internal Diseases, Sechenov First Moscow State Medical University, Rossolimo 11/5, Moscow, Russia, 119435
| | - Sergey Moiseev
- Tareev Clinic of Internal Diseases, Sechenov First Moscow State Medical University, Rossolimo 11/5, Moscow, Russia, 119435
| |
Collapse
|
157
|
Quintans JSS, Shanmugam S, Heimfarth L, Araújo AAS, Almeida JRGDS, Picot L, Quintans-Júnior LJ. Monoterpenes modulating cytokines - A review. Food Chem Toxicol 2018; 123:233-257. [PMID: 30389585 DOI: 10.1016/j.fct.2018.10.058] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/11/2018] [Accepted: 10/24/2018] [Indexed: 12/15/2022]
Abstract
Inflammatory response can be driven by cytokine production and is a pivotal target in the management of inflammatory diseases. Monoterpenes have shown that promising profile as agents which reduce the inflammatory process and also modulate the key chemical mediators of inflammation, such as pro and anti-inflammatory cytokines. The main interest focused on monoterpenes were to develop the analgesic and anti-inflammatory drugs. In this review, we summarized current knowledge on monoterpenes that produce anti-inflammatory effects by modulating the release of cytokines, as well as suggesting that which monoterpenoid molecules may be most effective in the treatment of inflammatory disease. Several different inflammatory markers were evaluated as a target of monoterpenes. The proinflammatory and anti-inflammatory cytokines were found TNF-α, IL-1β, IL-2, IL-5, IL-4, IL-6, IL-8, IL-10, IL-12 IL-13, IL-17A, IFNγ, TGF-β1 and IFN-γ. Our review found evidence that NF-κB and MAPK signaling are important pathways for the anti-inflammatory action of monoterpenes. We found 24 monoterpenes that modulate the production of cytokines, which appears to be the major pharmacological mechanism these compounds possess in relation to the attenuation of inflammatory response. Despite the compelling evidence supporting the anti-inflammatory effect of monoterpenes, further studies are necessary to fully explore their potential as anti-inflammatory compounds.
Collapse
Affiliation(s)
- Jullyana S S Quintans
- Laboratory of Neuroscience and Pharmacological Assays, Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Saravanan Shanmugam
- Laboratory of Neuroscience and Pharmacological Assays, Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Luana Heimfarth
- Laboratory of Neuroscience and Pharmacological Assays, Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | | | - Jackson R G da S Almeida
- Center for Studies and Research of Medicinal Plants (NEPLAME), Federal University of San Francisco Valley (UNIVASF), Petrolina, Pernambuco, Brazil
| | - Laurent Picot
- UMRi CNRS 7266 LIENSs, University of La Rochelle, 17042, La Rochelle, France
| | - Lucindo J Quintans-Júnior
- Laboratory of Neuroscience and Pharmacological Assays, Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.
| |
Collapse
|
158
|
A modified pectic polysaccharide from turmeric (Curcuma longa) with antiulcer effects via anti–secretary, mucoprotective and IL–10 mediated anti–inflammatory mechanisms. Int J Biol Macromol 2018; 118:864-880. [PMID: 29924982 DOI: 10.1016/j.ijbiomac.2018.06.053] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/03/2018] [Accepted: 06/11/2018] [Indexed: 12/15/2022]
|
159
|
LeVan TD, Romberger DJ, Siahpush M, Grimm BL, Ramos AK, Johansson PL, Michaud TL, Heires AJ, Wyatt TA, Poole JA. Relationship of systemic IL-10 levels with proinflammatory cytokine responsiveness and lung function in agriculture workers. Respir Res 2018; 19:166. [PMID: 30176916 PMCID: PMC6122449 DOI: 10.1186/s12931-018-0875-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 08/27/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Agriculture workers are exposed to microbial component- and particulate matter-enriched organic dust aerosols. Whereas it is clear that exposure to these aerosols can lead to lung inflammation, it is not known how inflammatory responses are resolved in some individuals while others develop chronic lung disease. Interleukin (IL)-10 is an immunomodulatory cytokine that is recognized as a potent anti-inflammatory and pro-resolving factor. The objective of this study was to determine whether there is a relationship of systemic IL-10 and proinflammatory responses and/or respiratory health effects in humans with prior agriculture exposure. METHODS This is a cross sectional study of 625 veterans with > 2 years of farming experience. Whole blood was stimulated with or without organic dust and measured for IL-6, TNFα and IL-10. Participants underwent spirometry and respiratory symptoms were assessed by questionnaire. RESULTS We found that baseline IL-10 concentration from the whole blood assay was inversely associated with ΔTNF-α (r = - 0.63) and ΔIL-6 (r = - 0.37) levels. Results remained highly significant in the linear regression model after adjusting for age, sex, BMI, race, education, smoking status, and white blood cell count (ΔTNF-α, p < 0.0001; ΔIL-6, p < 0.0001). We found no association between chronic cough (p = 0.18), chronic phlegm (p = 0.31) and chronic bronchitis (p = 0.06) and baseline IL-10 levels using univariate logistic regression models. However, we did find that higher FEV1/FVC was significantly associated with increased baseline IL-10 concentration. CONCLUSIONS Collectively, these studies support a potential role for IL-10 in modulating an inflammatory response and lung function in agriculture-exposed persons.
Collapse
Affiliation(s)
- Tricia D. LeVan
- College of Public Health, University of Nebraska Medical Center, Omaha, NE USA
- Veterans Affairs Nebraska Western Iowa Healthcare System, Omaha, NE USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE USA
- Department of Epidemiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Debra J. Romberger
- Veterans Affairs Nebraska Western Iowa Healthcare System, Omaha, NE USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE USA
| | - Mohammad Siahpush
- College of Public Health, University of Nebraska Medical Center, Omaha, NE USA
| | - Brandon L. Grimm
- College of Public Health, University of Nebraska Medical Center, Omaha, NE USA
| | - Athena K. Ramos
- College of Public Health, University of Nebraska Medical Center, Omaha, NE USA
| | - Patrik L. Johansson
- College of Public Health, University of Nebraska Medical Center, Omaha, NE USA
| | - Tzeyu L. Michaud
- College of Public Health, University of Nebraska Medical Center, Omaha, NE USA
| | - Art J. Heires
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE USA
| | - Todd A. Wyatt
- Veterans Affairs Nebraska Western Iowa Healthcare System, Omaha, NE USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE USA
- Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE USA
| | - Jill A. Poole
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE USA
| |
Collapse
|
160
|
Feng Q, Yao J, Zhou G, Xia W, Lyu J, Li X, Zhao T, Zhang G, Zhao N, Yang J. Quantitative Proteomic Analysis Reveals That Arctigenin Alleviates Concanavalin A-Induced Hepatitis Through Suppressing Immune System and Regulating Autophagy. Front Immunol 2018; 9:1881. [PMID: 30177931 PMCID: PMC6109684 DOI: 10.3389/fimmu.2018.01881] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 07/30/2018] [Indexed: 12/24/2022] Open
Abstract
Concanavalin A-induced autoimmune hepatitis is a well-established experimental model for immune-mediated liver injury. It has been widely used in the therapeutic studies of immune hepatitis. The in-depth analysis of dysregulated proteins from comparative proteomic results indicated that the activation of immune system resulted in the deregulation of autophagy. Follow-up studies validated that some immune related proteins, including Stat1, Pkr, Atg7, and Adrm1, were indeed upregulated. The accumulations of LC3B-II and p62 were confirmed by immunohistochemistry and Western blot analyses. Arctigenin pretreatment significantly alleviated the liver injury, as evidenced by biochemical and histopathological investigations, whose protective effects were comparable with Prednisone acetate and Cyclosporin A. Arctigenin pretreatment decreased the levels of IL-6 and IFN-γ, but increased the ones of IL-10. Next, the quantitative proteomic analysis demonstrated that ARC pretreatment suppressed the activation of immune system through the inhibition of IFN-γ signaling, when it downregulated the protein expressions of Stat1, P-Stat1, Pkr, P-Pkr, Bnip3, Beclin1, Atg7, LC3B, Adrm1, and p62. Meanwhile, Arctigenin pretreatment also reduced the gene expressions of Stat1, Pkr, and Atg7. These results suggested that Arctigenin alleviated autophagy as well as apoptosis through inhibiting IFN-γ/IL-6/Stat1 pathway and IL-6/Bnip3 pathway. In summary, the comparative proteomic analysis revealed that the activation of immune system led to Concanavalin A-induced hepatitis. Both autophagy and apoptosis had important clinical implications for the treatment of immune hepatitis. Arctigenin might exert great therapeutic potential in immune-mediated liver injury.
Collapse
Affiliation(s)
- Qin Feng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Jingchun Yao
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Ge Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenkai Xia
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Jingang Lyu
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Xin Li
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Tao Zhao
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Guimin Zhang
- Center for New Drug Pharmacological Research of Lunan Pharmaceutical Group, State Key Laboratory, Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China.,School of Pharmacy, Linyi University, Linyi, China
| | - Ningwei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Shimadzu Biomedical Research Laboratory, Shanghai, China
| | - Jie Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
161
|
Regulatory T cells as a new therapeutic target for atherosclerosis. Acta Pharmacol Sin 2018; 39:1249-1258. [PMID: 29323337 DOI: 10.1038/aps.2017.140] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/18/2017] [Indexed: 12/21/2022]
Abstract
Atherosclerosis is an autoimmune disease caused by self- and non-self-antigens contributing to excessive activation of T and B cell immune responses. These responses further aggravate vascular infiammation and promote progression of atherosclerosis and vulnerability to plaques via releasing pro-infiammatory cytokines. Regulatory T cells (Tregs) as the major immunoregulatory cells, in particular, induce and maintain immune homeostasis and tolerance by suppressing the immune responses of various cells such as T and B cells, natural killer (NK) cells, monocytes, and dendritic cells (DCs), as well as by secreting inhibitory cytokines interleukin (IL)-10, IL-35 and transcription growth factor β (TGF-β) in both physiological and pathological states. Numerous evidence demonstrates that reduced numbers and dysfunction of Treg may be involveved in atherosclerosis pathogenesis. Increasing or restoring the numbers and improving the immunosuppressive capacity of Tregs may serve as a fundamental immunotherapy to treat atherosclerotic cardiovascular diseases. In this article, we briefiy present current knowledge of Treg subsets, summarize the relationship between Tregs and atherosclerosis development, and discuss the possibilities of regulating Tregs for prevention of atherosclerosis pathogenesis and enhancement of plaque stability. Although the exact molecular mechanisms of Treg-mediated protection against atherosclerosis remain to be elucidated, the strategies for targeting the regulation of Tregs may provide specific and significant approaches for the prevention and treatment of atherosclerotic cardiovascular diseases.
Collapse
|
162
|
Luan J, Zhang K, Yang P, Zhang Y, Feng F, Zhu YM, Zhu P, Chen ZN. The combination of FK506 and an anti-CD147 mAb exerts potential therapeutic effects on a mouse model of collagen-induced arthritis. Mol Immunol 2018; 101:1-9. [PMID: 29852454 DOI: 10.1016/j.molimm.2018.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 05/02/2018] [Accepted: 05/14/2018] [Indexed: 01/16/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease, and excessive T lymphocyte activation plays a critical role in the development of inflammation. CD147 is an antigen related to T cell activation, CD147 blockade exerts beneficial effects on RA. FK506, also known as tacrolimus, exerts strong immunosuppressive effects by inhibiting T cell activation. In this study, RL73 (an anti-mouse CD147 functional-grade purified antibody) and FK506 were co-administered to mice with collagen-induced arthritis (CIA). As expected, the combination of these two drugs produced superior therapeutic effects than either drug alone and enabled the administration of each drug at a lower dose. Moreover, joint damage and destruction were significantly improved in mice injected with both FK506 and RL73 compared with mice injected with either agent alone. These effects might have been observed because the proportions of CD4 + T and CD8 + T cells in the mouse spleen of the combination regimen were clearly decreased compared with each monotherapy. In addition, the proportions of Th2 subsets in the mouse spleen and peripheral blood were clearly increased, and the serum levels of the cytokines interleukin 4 (IL-4) and IL-10 were markedly increased in mice treated with the combination therapy compared with the other groups of mice. The splenic total number of T lymphocytes also showed that the inhibition of T lymphocytes was the most obvious in the combined treatment group. Based on the results from the present study, combining FK506 and the anti-CD147 mAb might be a new practical therapeutic option for the treatment of RA.
Collapse
Affiliation(s)
- Jing Luan
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China; Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University , Xi'an, 710032, China; Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Kui Zhang
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China; Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Peng Yang
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China; Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University , Xi'an, 710032, China
| | - Yang Zhang
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China; Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University , Xi'an, 710032, China.
| | - Fei Feng
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China; Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University , Xi'an, 710032, China
| | - Yu-Meng Zhu
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China; Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University , Xi'an, 710032, China
| | - Ping Zhu
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China; Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Zhi-Nan Chen
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China; Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University , Xi'an, 710032, China.
| |
Collapse
|
163
|
Luo Y, McShan DL, Matuszak MM, Ray D, Lawrence TS, Jolly S, Kong FM, Ten Haken RK, Naqa IE. A multiobjective Bayesian networks approach for joint prediction of tumor local control and radiation pneumonitis in nonsmall-cell lung cancer (NSCLC) for response-adapted radiotherapy. Med Phys 2018; 45:10.1002/mp.13029. [PMID: 29862533 PMCID: PMC6279602 DOI: 10.1002/mp.13029] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/28/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Individualization of therapeutic outcomes in NSCLC radiotherapy is likely to be compromised by the lack of proper balance of biophysical factors affecting both tumor local control (LC) and side effects such as radiation pneumonitis (RP), which are likely to be intertwined. Here, we compare the performance of separate and joint outcomes predictions for response-adapted personalized treatment planning. METHODS A total of 118 NSCLC patients treated on prospective protocols with 32 cases of local progression and 20 cases of RP grade 2 or higher (RP2) were studied. Sixty-eight patients with 297 features before and during radiotherapy were used for discovery and 50 patients were reserved for independent testing. A multiobjective Bayesian network (MO-BN) approach was developed to identify important features for joint LC/RP2 prediction using extended Markov blankets as inputs to develop a BN predictive structure. Cross-validation (CV) was used to guide the MO-BN structure learning. Area under the free-response receiver operating characteristic (AU-FROC) curve was used to evaluate joint prediction performance. RESULTS Important features including single nucleotide polymorphisms (SNPs), micro RNAs, pretreatment cytokines, pretreatment PET radiomics together with lung and tumor gEUDs were selected and their biophysical inter-relationships with radiation outcomes (LC and RP2) were identified in a pretreatment MO-BN. The joint LC/RP2 prediction yielded an AU-FROC of 0.80 (95% CI: 0.70-0.86) upon internal CV. This improved to 0.85 (0.75-0.91) with additional two SNPs, changes in one cytokine and two radiomics PET image features through the course of radiotherapy in a during-treatment MO-BN. This MO-BN model outperformed combined single-objective Bayesian networks (SO-BNs) during-treatment [0.78 (0.67-0.84)]. AU-FROC values in the evaluation of the MO-BN and individual SO-BNs on the testing dataset were 0.77 and 0.68 for pretreatment, and 0.79 and 0.71 for during-treatment, respectively. CONCLUSIONS MO-BNs can reveal possible biophysical cross-talks between competing radiotherapy clinical endpoints. The prediction is improved by providing additional during-treatment information. The developed MO-BNs can be an important component of decision support systems for personalized response-adapted radiotherapy.
Collapse
Affiliation(s)
- Yi Luo
- Department of Radiation Oncology, the University of Michigan, Ann Arbor, Michigan, 48103 United States
| | - Daniel L. McShan
- Department of Radiation Oncology, the University of Michigan, Ann Arbor, Michigan, 48103 United States
| | - Martha M. Matuszak
- Department of Radiation Oncology, the University of Michigan, Ann Arbor, Michigan, 48103 United States
| | - Dipankar Ray
- Department of Radiation Oncology, the University of Michigan, Ann Arbor, Michigan, 48103 United States
| | - Theodore S. Lawrence
- Department of Radiation Oncology, the University of Michigan, Ann Arbor, Michigan, 48103 United States
| | - Shruti Jolly
- Department of Radiation Oncology, the University of Michigan, Ann Arbor, Michigan, 48103 United States
| | - Feng-Ming Kong
- Department of Radiation Oncology, Indiana University, Indianapolis, Indiana, 46202 United States
| | - Randall K. Ten Haken
- Department of Radiation Oncology, the University of Michigan, Ann Arbor, Michigan, 48103 United States
| | - Issam El Naqa
- Department of Radiation Oncology, the University of Michigan, Ann Arbor, Michigan, 48103 United States
| |
Collapse
|
164
|
Cambier L, Giani JF, Liu W, Ijichi T, Echavez AK, Valle J, Marbán E. Angiotensin II-Induced End-Organ Damage in Mice Is Attenuated by Human Exosomes and by an Exosomal Y RNA Fragment. Hypertension 2018; 72:370-380. [PMID: 29866742 DOI: 10.1161/hypertensionaha.118.11239] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 04/06/2018] [Accepted: 04/16/2018] [Indexed: 01/06/2023]
Abstract
Hypertension often leads to cardiovascular disease and kidney dysfunction. Exosomes secreted from cardiosphere-derived cells (CDC-exo) and their most abundant small RNA constituent, the Y RNA fragment EV-YF1, exert therapeutic benefits after myocardial infarction. Here, we investigated the effects of CDC-exo and EV-YF1, each administered individually, in a model of cardiac hypertrophy and kidney injury induced by chronic infusion of Ang (angiotensin) II. After 2 weeks of Ang II, multiple doses of CDC-exo or EV-YF1 were administered retro-orbitally. Ang II infusion induced an elevation in systolic blood pressure that was not affected by CDC-exo or EV-YF1. Echocardiography confirmed that Ang II infusion led to cardiac hypertrophy. CDC-exo and EV-YF1 both attenuated cardiac hypertrophy and reduced cardiac inflammation and fibrosis. In addition, both CDC-exo and EV-YF1 improved kidney function and diminished renal inflammation and fibrosis. The beneficial effects of CDC-exo and EV-YF1 were associated with changes in the expression of the anti-inflammatory cytokine IL (interleukin)-10 in plasma, heart, spleen, and kidney. In summary, infusions of CDC-exo or EV-YF1 attenuated cardiac hypertrophy and renal injury induced by Ang II infusion, without affecting blood pressure, in association with altered IL-10 expression. Exosomes and their defined noncoding RNA contents may represent potential new therapeutic approaches for hypertension-associated cardiovascular and renal damage.
Collapse
Affiliation(s)
- Linda Cambier
- From the Smidt Heart Institute (L.C., W.L., T.I., A.K.E., J.V., E.M.).,Department of Biomedical Sciences (L.C., J.F.G.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jorge F Giani
- Department of Biomedical Sciences (L.C., J.F.G.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Weixin Liu
- From the Smidt Heart Institute (L.C., W.L., T.I., A.K.E., J.V., E.M.)
| | - Takeshi Ijichi
- From the Smidt Heart Institute (L.C., W.L., T.I., A.K.E., J.V., E.M.)
| | - Antonio K Echavez
- From the Smidt Heart Institute (L.C., W.L., T.I., A.K.E., J.V., E.M.)
| | - Jackelyn Valle
- From the Smidt Heart Institute (L.C., W.L., T.I., A.K.E., J.V., E.M.)
| | - Eduardo Marbán
- From the Smidt Heart Institute (L.C., W.L., T.I., A.K.E., J.V., E.M.)
| |
Collapse
|
165
|
White JR, Phillips F, Monaghan T, Fateen W, Samuel S, Ghosh S, Moran GW. Review article: novel oral-targeted therapies in inflammatory bowel disease. Aliment Pharmacol Ther 2018; 47:1610-1622. [PMID: 29672874 DOI: 10.1111/apt.14669] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 12/12/2017] [Accepted: 03/25/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND There is a great unmet clinical need for efficacious, tolerable, economical and orally administrated drugs for the treatment of inflammatory bowel disease (IBD). New therapeutic avenues have become possible including the development of medications that target specific genetic pathways found to be relevant in other immune mediated diseases. AIMS To provide an overview of recent clinical trials for new generation oral targeted medications that may have a future role in IBD management. METHODS Pubmed and Medline searches were performed up to 1 March 2018 using keywords: "IBD", "UC", "CD", "inflammatory bowel disease" "ulcerative colitis", "Crohn's disease" in combination with "phase", "study", "trial" and "oral". A manual search of the clinical trial register, article reference lists, abstracts from meetings of Digestive Disease Week, United European Gastroenterology Week and ECCO congress were also conducted. RESULTS In randomised controlled trials primary efficacy endpoints were met for tofacitinib (JAK 1/3 inhibitor-phase III), upadacitinib (JAK 1 inhibitor-phase II) and AJM300 (α4-integrin antagonist-phase II) in ulcerative colitis. Ozanimod (S1P receptor agonist-phase II) also demonstrated clinical remission. For Crohn's disease, filgotinib (JAK1 inhibitor-phase II) met primary endpoints and laquinimod (quinolone-3-carboxide small molecule-phase II) was also efficacious. Trials using mongersen (SMAD7 inhibitor) and vidofludimus (dihydroorotate dehydrogenase inhibitor) have been halted. CONCLUSIONS This is potentially the start of an exciting new era in which multiple therapeutic options are at the disposal of physicians to treat IBD on an individualised basis. Head-to-head studies with existing treatments and longer term safety data are needed for this to be possible.
Collapse
Affiliation(s)
- J R White
- NIHR Biomedical Research Centre in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust, The University of Nottingham, Nottingham, UK
| | - F Phillips
- NIHR Biomedical Research Centre in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust, The University of Nottingham, Nottingham, UK
| | - T Monaghan
- NIHR Biomedical Research Centre in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust, The University of Nottingham, Nottingham, UK
| | - W Fateen
- NIHR Biomedical Research Centre in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust, The University of Nottingham, Nottingham, UK
| | - S Samuel
- NIHR Biomedical Research Centre in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust, The University of Nottingham, Nottingham, UK
| | - S Ghosh
- NIHR Biomedical Research Centre, Institute of Translational Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - G W Moran
- NIHR Biomedical Research Centre in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust, The University of Nottingham, Nottingham, UK
| |
Collapse
|
166
|
Pellerin L, Chen P, Gregori S, Hernandez-Hoyos G, Bacchetta R, Roncarolo MG. APVO210: A Bispecific Anti-CD86-IL-10 Fusion Protein (ADAPTIR™) to Induce Antigen-Specific T Regulatory Type 1 Cells. Front Immunol 2018; 9:881. [PMID: 29887861 PMCID: PMC5980965 DOI: 10.3389/fimmu.2018.00881] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/09/2018] [Indexed: 12/30/2022] Open
Abstract
IL-10 is a potent immunosuppressive cytokine that promotes the differentiation of tolerogenic dendritic cells (DC-10), and the subsequent induction of antigen-specific T regulatory type 1 (Tr1) cells, which suppress immune responses. However, IL-10 acts on multiple cell types and its effects are not solely inhibitory, therefore, limiting its use as immunomodulant. APVO210 is a bispecific fusion protein composed of an anti-CD86 antibody fused with monomeric IL-10 (ADAPTIR™ from Aptevo Therapeutics). APVO210 specifically induces IL-10R signaling in CD86+ antigen-presenting cells, but not in T and B cells. In this study, we tested whether APVO210 promotes the differentiation of tolerogenic DC-10 and the differentiation of antigen-specific CD4+ Tr1 cells in vitro. We compared the effect of APVO210 with that of recombinant human (rh) IL-10 on the in vitro differentiation of DC-10, induction of alloantigen-specific anergic CD4+ T cells, enrichment in CD49b+LAG3+ Tr1 cells mediating antigen-specific suppression, and stability upon exposure to inflammatory cytokines. APVO210 induced the differentiation of tolerogenic DC (DC-A210) that produced high levels of IL-10, expressed CD86, HLA-G, and intermediate levels of CD14 and CD16. These DC-A210 induced alloantigen-specific anergic T-cell cultures (T-alloA210) that were enriched in CD49b+ LAG3+ Tr1 cells, produced high levels of IL-10, and had suppressive properties. The phenotype and high IL-10 production by DC-A210, and the alloantigen-specific anergy of T-alloA210 were preserved upon exposure to the inflammatory cytokines IL-1β, IL-6, and TNF-α. The effects of APVO210 were comparable to that of dimeric rh IL-10. In conclusion, our data demonstrate that APVO210 drives the differentiation of tolerogenic DC and functional alloantigen-specific Tr1 cells in vitro. Since APVO210 specifically targets CD86+ cells, we hypothesize that it will specifically target CD86+ DC to induce Tr1 cells in vivo, and mediate antigen-specific immunological tolerance by induction of tolerogenic DC and Tr1 cells.
Collapse
Affiliation(s)
- Laurence Pellerin
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| | - Ping Chen
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Rosa Bacchetta
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| | - Maria Grazia Roncarolo
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
167
|
Dou J, Bennett MR. Synthetic Biology and the Gut Microbiome. Biotechnol J 2018; 13:e1700159. [PMID: 28976641 PMCID: PMC5882594 DOI: 10.1002/biot.201700159] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/23/2017] [Indexed: 12/19/2022]
Abstract
The gut microbiome plays a crucial role in maintaining human health. Functions performed by gastrointestinal microbes range from regulating metabolism to modulating immune and nervous system development. Scientists have attempted to exploit this importance through the development of engineered probiotics that are capable of producing and delivering small molecule therapeutics within the gut. However, existing synthetic probiotics are simplistic and fail to replicate the complexity and adaptability of native homeostatic mechanisms. In this review, the ways in which the tools and approaches of synthetic biology have been applied to improve the efficacy of therapeutic probiotics, and the ways in which they might be applied in the future is discussed. Simple devices, such as a bistable switches and integrase memory arrays, have been successfully implemented in the mammalian gut, and models for targeted delivery in this environment have also been developed. In the future, it will be necessary to introduce concepts such as logic-gating and biocontainment mechanisms into synthetic probiotics, as well as to expand the collection of relevant biosensors. Ideally, this will bring us closer to a reality in which engineered therapeutic microbes will be able to accurately diagnose and effectively respond to a variety of disease states.
Collapse
Affiliation(s)
- Jennifer Dou
- Department of Biosciences, Rice University, Houston, TX 77005
| | - Matthew R. Bennett
- Department of Biosciences, Rice University, Houston, TX 77005
- Department of Bioengineering, Rice University, Houston, TX 77005
| |
Collapse
|
168
|
Nociceptor interleukin 10 receptor 1 is critical for muscle analgesia induced by repeated bouts of eccentric exercise in the rat. Pain 2018. [PMID: 28628078 DOI: 10.1097/j.pain.0000000000000936] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Delayed-onset muscle soreness is typically observed after strenuous or unaccustomed eccentric exercise. Soon after recovery, blunted muscle soreness is observed on repeated eccentric exercise, a phenomenon known as repeated bout effect (RBE). Although regular physical activity decreases muscle hyperalgesia, likely because of increased production of the anti-inflammatory cytokine interleukin-10 (IL-10) in the skeletal muscle, whether IL-10 also contributes to the antinociceptive effect of RBE is unknown. Furthermore, whether IL-10 attenuates muscle hyperalgesia by acting on muscle nociceptors remains to be established. Here, we explored the hypothesis that blunted muscle nociception observed in RBE depends on a local effect of IL-10, acting on IL-10 receptor 1 (IL-10R1) expressed by muscle nociceptors. Results show that after a second bout of eccentric exercise, rats exhibited decreased muscle hyperalgesia, indicative of RBE, and increased expression of IL-10 in the exercised gastrocnemius muscle. Although knockdown of IL-10R1 protein in nociceptors innervating the gastrocnemius muscle by intrathecal antisense oligodeoxynucleotide did not change nociceptive threshold in naive rats, it unveiled latent muscle hyperalgesia in rats submitted to eccentric exercise 12 days ago. Furthermore, antisense also prevented the reduction of muscle hyperalgesia observed after a second bout of eccentric exercise. These data indicate that recovery of nociceptive threshold after eccentric exercise and RBE-induced analgesia depend on a local effect of IL-10, acting on its canonical receptor in muscle nociceptors.
Collapse
|
169
|
Granulocyte-macrophage colony-stimulating factor negatively regulates early IL-10-mediated responses. Future Sci OA 2018; 4:FSO288. [PMID: 29682323 PMCID: PMC5905582 DOI: 10.4155/fsoa-2017-0133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/09/2018] [Indexed: 01/10/2023] Open
Abstract
Aim: Treatment of inflammatory disorders relies on the intervention in immune responses thereby restoring homeostasis. IL-10 is a cytokine with therapeutic potential, but until now has not been as successful as previously anticipated. A reason for this may be that IL-10 responsiveness depends on the environment of the inflamed tissue. In this study we investigated whether GM-CSF is able to influence IL-10-mediated responses. Methodology: Dendritic cells and macrophages were differentiated from mouse bone marrow and treated or depleted from GM-CSF prior to analyze their response to IL-10. Activity was assessed by measuring cytokine expression upon lipopolysaccharide stimulation, IL-10-induced signaling and down-stream gene expression. Conclusion: This study describes that GM-CSF negatively regulates IL-10-mediated responses. Over the last couple of decades inflammatory disorders, like autoimmune disease and allergies, are becoming more prevalent in the western world. These inflammatory disorders are characterized by uncontrolled immune responses against harmless antigens or commensal bacteria. Treatment of these diseases relies on the intervention in inflammatory responses and thereby restoring the balance of the immune system. One approach used in the clinic to balance the immune system is by treating patients with molecules used by our own immune system to suppress inflammation, such as IL-10. However, treatment with IL-10 has not been as successful as previously anticipated. In this study we show that a particular signaling molecule of the immune system that contributes to inflammation negatively affects the response of immune cells toward IL-10 and thereby could contribute to the low efficacy of IL-10 treatment.
Collapse
|
170
|
Ahluwalia B, Moraes L, Magnusson MK, Öhman L. Immunopathogenesis of inflammatory bowel disease and mechanisms of biological therapies. Scand J Gastroenterol 2018. [PMID: 29523023 DOI: 10.1080/00365521.2018.1447597] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract with a multifactorial pathophysiology. Full comprehension of IBD pathology is still out of reach and, therefore, treatment is far from ideal. Nevertheless, components involved in IBD pathogenesis including environmental, genetic, microbial, and immunological factors are continuously being investigated and the improved knowledge contributes to the development of new therapies. In this article we review the aspects of the immunopathogenesis of IBD, with focus on mucosal immunity, and discuss mechanisms of action for current and emerging biological therapies.
Collapse
Affiliation(s)
- Bani Ahluwalia
- a Department of Microbiology and Immunology , University of Gothenburg, Institute for Biomedicine, Sahlgrenska Academy , Gothenburg , Sweden.,b Research Unit , Calmino Group AB , Gothenburg , Sweden
| | - Luiza Moraes
- a Department of Microbiology and Immunology , University of Gothenburg, Institute for Biomedicine, Sahlgrenska Academy , Gothenburg , Sweden
| | - Maria K Magnusson
- a Department of Microbiology and Immunology , University of Gothenburg, Institute for Biomedicine, Sahlgrenska Academy , Gothenburg , Sweden
| | - Lena Öhman
- a Department of Microbiology and Immunology , University of Gothenburg, Institute for Biomedicine, Sahlgrenska Academy , Gothenburg , Sweden.,c Department of Internal Medicine and Clinical Nutrition , University of Gothenburg, Institute for Medicine, Sahlgrenska Academy , Gothenburg , Sweden
| |
Collapse
|
171
|
Agrawal U, Kumari N, Mishra AK, Vasudeva P, Kumar A, Mohanty NK, Saxena S. Immune signature of urothelial cancer associated with grade, recurrence, and invasion. Urol Oncol 2018; 34:418.e17-26. [PMID: 27554791 DOI: 10.1016/j.urolonc.2016.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 04/19/2016] [Accepted: 04/23/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND Urothelial carcinoma (UC) is one of most common genitourinary malignancy and the spectrum of disease ranges from in situ lesions to muscle-invasive cancers. The non-muscle-invasive lesions have tendency to recur or progress to muscle-invasive disease. The study of the immune profile may identify immune determinants associated with high-grade, recurrence, and invasion in patients with UC. METHODS Pathway-focused RT(2) profiler arrays were used to screen patients with UC for dysregulation of candidate genes of Th1-Th2-Th3 and NFκB pathways, which were then validated by real-time polymerase chain reaction on tumor samples and correlated with grade, recurrence, and invasion of tumors to identify their role in predicting behavior of the tumor. The cytokines found associated with recurrence were then validated in urine of patients with UC. RESULTS IFNγ, IL2, IL4, IL10, IL17, CCL7, CTLA4, and SPP1 of the cytokine pathway and TLR4, TLR3, RELA, NFκB1, and MYD88 of the NFκB pathway were found differentially expressed in patients with urothelial cancer by array and quantative real-time polymerase chain reaction. Among these, IL10 and SPP1 were found consistently up-regulated in high-grade, invasive, and recurrent cases and up-regulated IL10 and CTLA4 were found associated with a short recurrence-free survival time (P = 0.001 and P = 0.065). Urinary IL10 concentration was significantly higher in both patients with cancer and cystitis compared with healthy controls, but the difference in concentration between patients with cancer and cystitis patients was not statistically significant. However, urinary CTLA4 concentrations were found to be significantly higher in urothelial cancer patients compared with healthy controls and cystitis cases and found to be associated with poor recurrence-free survival. CONCLUSION The study indicates that high urinary CTLA4 concentration raises the index of suspicion of recurrence in a known case of urothelial cancer and may be used as a surveillance marker.
Collapse
Affiliation(s)
- Usha Agrawal
- National Institute of Pathology, Indian Council of Medical Research, New Delhi, India; Symbiosis International University, Lavale, Pune, India
| | - Nitu Kumari
- National Institute of Pathology, Indian Council of Medical Research, New Delhi, India; BITS, Pilani, Rajasthan, India
| | | | - Pawan Vasudeva
- Deptartment of Urology, VMMC and Safdarjung Hospital, New Delhi, India
| | - Anup Kumar
- Deptartment of Urology, VMMC and Safdarjung Hospital, New Delhi, India
| | | | - Sunita Saxena
- National Institute of Pathology, Indian Council of Medical Research, New Delhi, India.
| |
Collapse
|
172
|
Intestinal Immunomodulatory Cells (T Lymphocytes): A Bridge between Gut Microbiota and Diabetes. Mediators Inflamm 2018; 2018:9830939. [PMID: 29713241 PMCID: PMC5866888 DOI: 10.1155/2018/9830939] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/10/2018] [Accepted: 01/28/2018] [Indexed: 12/20/2022] Open
Abstract
Diabetes mellitus (DM) is one of the most familiar chronic diseases threatening human health. Recent studies have shown that the development of diabetes is closely related to an imbalance of the gut microbiota. Accordingly, there is increasing interest in how changes in the gut microbiota affect diabetes and its underlying mechanisms. Immunomodulatory cells play important roles in maintaining the normal functioning of the human immune system and in maintaining homeostasis. Intestinal immunomodulatory cells (IICs) are located in the intestinal mucosa and are regarded as an intermediary by which the gut microbiota affects physiological and pathological properties. Diabetes can be regulated by IICs, which act as a bridge linking the gut microbiota and DM. Understanding this bridge role of IICs may clarify the mechanisms by which the gut microbiota contributes to DM. Based on recent research, we summarize this process, thereby providing a basis for further studies of diabetes and other similar immune-related diseases.
Collapse
|
173
|
Smith CJ, Allard DE, Wang Y, Howard JF, Montgomery SA, Su MA. IL-10 Paradoxically Promotes Autoimmune Neuropathy through S1PR1-Dependent CD4 + T Cell Migration. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:1580-1592. [PMID: 29367208 PMCID: PMC5821539 DOI: 10.4049/jimmunol.1701280] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/28/2017] [Indexed: 01/13/2023]
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is a debilitating condition caused by autoimmune demyelination of peripheral nerves. CIDP is associated with increased IL-10, a cytokine with well-described anti-inflammatory effects. However, the role of IL-10 in CIDP is unclear. In this study, we demonstrate that IL-10 paradoxically exacerbates autoimmunity against peripheral nerves. In IL-10-deficient mice, protection from neuropathy was associated with an accrual of highly activated CD4+ T cells in draining lymph nodes and absence of infiltrating immune cells in peripheral nerves. Accumulated CD4+ T cells in draining lymph nodes of IL-10-deficient mice expressed lower sphingosine-1-phosphate receptor 1 (S1pr1), a protein important in lymphocyte egress. Additionally, IL-10 stimulation in vitro induced S1pr1 expression in lymph node cells in a STAT3-dependent manner. Together, these results delineate a novel mechanism in which IL-10-induced STAT3 increases S1pr1 expression and CD4+ T cell migration to accelerate T cell-mediated destruction of peripheral nerves.
Collapse
Affiliation(s)
- Collin-Jamal Smith
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Denise E Allard
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Yan Wang
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - James F Howard
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Maureen A Su
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599;
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
174
|
Guimaraes TS, da Rocha LA, Becari C, Piccinato CE, Joviliano RD, Ribeiro MS, Joviliano EE. The Role of Interleukins and Inflammatory Markers in the Early Restenosis of Covered Stents in the Femoropopliteal Arterial Segment. Ann Vasc Surg 2018; 50:88-95. [PMID: 29481941 DOI: 10.1016/j.avsg.2017.11.064] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/22/2017] [Accepted: 11/26/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND The objective of this study was to evaluate the relationship between inflammatory markers, such as interleukin (IL)-1β, IL-6, IL-8, IL-10, tumor necrosis factor α (TNF-α), transforming growth factor β (TGF-β), and highly sensitive C-reactive protein, and the development of arterial restenosis 6 months after femoropopliteal percutaneous transluminal angioplasty (PTA) with covered stent implantation. METHODS We recruited 27 patients of a tertiary hospital in Brazil who were treated with covered stents for atherosclerotic peripheral arterial disease. Serum samples were collected before stent implantation, then 24 hr later, and 6 months after the procedure. RESULTS At 6-month follow-up, 4 patients (15%) presented restenosis. IL1- β, IL-6, IL-8, and TNF-α levels showed a statistically significant reduction after both 24 hr and 6 months compared with pretreatment levels (P < 0.01). There were increased levels of IL-10 and TGF-β both 24 hr and 6 months after PTA and stenting compared with pretreatment levels (P < 0.01). None of the cytokines studied were correlated with restenosis. CONCLUSIONS This study demonstrated a significant increase in anti-inflammatory TGF-β and IL-10 and a decrease in proinflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α 6 months after the procedure, but no inflammatory marker was independently identified as a risk factor for in-stent restenosis.
Collapse
Affiliation(s)
- Thiago Silva Guimaraes
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Laura Andrade da Rocha
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Universidade Federal de Uberlandia, Department of Surgery, Uberlandia, Minas Gerais, Brazil.
| | - Christiane Becari
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carlos Eli Piccinato
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Renata Dellalibera Joviliano
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Mauricio Serra Ribeiro
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Edwaldo Edner Joviliano
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
175
|
Low HB, Png CW, Li C, Wang DY, Wong SBJ, Zhang Y. Monocyte-derived factors including PLA2G7 induced by macrophage-nasopharyngeal carcinoma cell interaction promote tumor cell invasiveness. Oncotarget 2018; 7:55473-55490. [PMID: 27487154 PMCID: PMC5342430 DOI: 10.18632/oncotarget.10980] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/29/2016] [Indexed: 02/07/2023] Open
Abstract
The non-keratinizing undifferentiated subtype of nasopharyngeal carcinoma (NPC) is a malignancy characterized by an intimate relationship between neoplastic cells and a non-neoplastic lymphoid component. Tumor-associated macrophages (TAMs) foster tumor progression through production of soluble mediators that support proliferation, angiogenesis, survival and invasion of malignant cells. However, the role of macrophages in the progression of NPC remains poorly understood. This study aims to investigate the functional and phenotypic changes that occur to macrophages in macrophage-NPC cell co-culture systems, and how these changes influence tumor cells. We found that monocytes, including THP-1 cells and primary human monocytes, co-cultured with C666-1 NPC cells upregulate expression of pro-inflammatory cytokines at the early stages, followed by the induction of metastasis-related genes and interferon-stimulated genes at the later stage of coculture, indicating that TAMs are “educated” by NPC cells for cancer progression. Importantly, the induction of these factors from the TAMs was also found to enhance the migratory capabilities of the NPC cells. We have also identified one of these macrophage-derived factor, phospholipase A2 Group 7 (PLA2G7), to be important in regulating tumor cell migration and a novel tumor-promoting factor in NPC. Further studies to characterize the role of PLA2G7 in tumor metastasis may help determine its potential as a therapeutic target in NPC.
Collapse
Affiliation(s)
- Heng Boon Low
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.,Immunology Programme, The Life Science Institute, National University of Singapore, Singapore 117597, Singapore
| | - Chin Wen Png
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.,Immunology Programme, The Life Science Institute, National University of Singapore, Singapore 117597, Singapore
| | - Chunwei Li
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Soon Boon Justin Wong
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.,Immunology Programme, The Life Science Institute, National University of Singapore, Singapore 117597, Singapore.,Department of Pathology, National University Hospital, Singapore 119074, Singapore
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.,Immunology Programme, The Life Science Institute, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
176
|
Pandey M, Awasthi S, Singh U, Mahdi AA. Association of IL-10 Gene Polymorphism (-819C > T, -592C > A and -1082G > A) with Preterm Birth. Indian J Pediatr 2018; 85:93-101. [PMID: 29147889 DOI: 10.1007/s12098-017-2496-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 09/14/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To analyze the association of IL-10 gene and its polymorphisms with preterm birth (PTB). METHODS Five hundred and fifty nine women with term birth and 559 with preterm birth were recruited from Lucknow, India. Genetic association analysis was conducted between cases and controls. Subjects recruited as cases were women (aged between 18-40 y) with singleton delivery before 37 wk of gestation and controls were with delivery after or on 37 wk. The genotyping was performed for rs1800871, rs1800872 and rs1800896 for assessing the allelic distribution, haplotypic association and linkage disequilibrium analysis. IL-10mRNA levels were evaluated by real time quantitative polymerase chain reaction (PCR) method. RESULTS The risk of PTB was found significant in women carrying IL-10 (-1082) GA genotype [OR=1.72(1.7-2.5), p=0.006]. The haplotypic analysis of studied polymorphisms for rs1800871, rs1800872 and rs1800896 depicted the association of ATA (p=0.02) and ATC (p=0.01) haplotypes with PTB. The IL-10 mRNA levels were significantly lower in cases (p=0.05). CONCLUSIONS IL-10 marks a protective impact in the inflammatory pathway of PTB.
Collapse
Affiliation(s)
- Monika Pandey
- Department of Pediatrics, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Shally Awasthi
- Department of Pediatrics, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India.
| | - Urmila Singh
- Department of Obstetrics and Gynecology, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Abbas Ali Mahdi
- Department of Biochemistry, King George's Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
177
|
Morales V, Soto-Ortiz L. Modeling Macrophage Polarization and Its Effect on Cancer Treatment Success. ACTA ACUST UNITED AC 2018; 8:36-80. [PMID: 35847834 PMCID: PMC9286492 DOI: 10.4236/oji.2018.82004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Positive feedback loops drive immune cell polarization toward a pro-tumor phenotype that accentuates immunosuppression and tumor angiogenesis. This phenotypic switch leads to the escape of cancer cells from immune destruction. These positive feedback loops are generated by cytokines such as TGF-β, Interleukin-10 and Interleukin-4, which are responsible for the polarization of monocytes and M1 macrophages into pro-tumor M2 macrophages, and the polarization of naive helper T cells intopro-tumor Th2 cells. In this article, we present a deterministic ordinary differential equation (ODE) model that includes key cellular interactions and cytokine signaling pathways that lead to immune cell polarization in the tumor microenvironment. The model was used to simulate various cancer treatments in silico. We identified combination therapies that consist of M1 macrophages or Th1 helper cells, coupled with an anti-angiogenic treatment, that are robust with respect to immune response strength, initial tumor size and treatment resistance. We also identified IL-4 and IL-10 as the targets that should be neutralized in order to make these combination treatments robust with respect to immune cell polarization. The model simulations confirmed a hypothesis based on published experimental evidence that a polarization into the M1 and Th1 phenotypes to increase the M1-to-M2 and Th1-to-Th2 ratios plays a significant role in treatment success. Our results highlight the importance of immune cell reprogramming as a viable strategy to eradicate a highly vascularized tumor when the strength of the immune response is characteristically weak and cell polarization to the pro-tumor phenotype has occurred.
Collapse
Affiliation(s)
- Valentin Morales
- Department of Engineering and Technologies, East Los Angeles College, Monterey Park, USA
| | - Luis Soto-Ortiz
- Department of Mathematics, East Los Angeles College, Monterey Park, USA
| |
Collapse
|
178
|
Li B, Concepcion K, Meng X, Zhang L. Brain-immune interactions in perinatal hypoxic-ischemic brain injury. Prog Neurobiol 2017; 159:50-68. [PMID: 29111451 PMCID: PMC5831511 DOI: 10.1016/j.pneurobio.2017.10.006] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/26/2017] [Indexed: 01/07/2023]
Abstract
Perinatal hypoxia-ischemia remains the primary cause of acute neonatal brain injury, leading to a high mortality rate and long-term neurological deficits, such as behavioral, social, attentional, cognitive and functional motor deficits. An ever-increasing body of evidence shows that the immune response to acute cerebral hypoxia-ischemia is a major contributor to the pathophysiology of neonatal brain injury. Hypoxia-ischemia provokes an intravascular inflammatory cascade that is further augmented by the activation of resident immune cells and the cerebral infiltration of peripheral immune cells response to cellular damages in the brain parenchyma. This prolonged and/or inappropriate neuroinflammation leads to secondary brain tissue injury. Yet, the long-term effects of immune activation, especially the adaptive immune response, on the hypoxic-ischemic brain still remain unclear. The focus of this review is to summarize recent advances in the understanding of post-hypoxic-ischemic neuroinflammation triggered by the innate and adaptive immune responses and to discuss how these mechanisms modulate the brain vulnerability to injury. A greater understanding of the reciprocal interactions between the hypoxic-ischemic brain and the immune system will open new avenues for potential immunomodulatory therapy in the treatment of neonatal brain injury.
Collapse
Affiliation(s)
- Bo Li
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Katherine Concepcion
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Xianmei Meng
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
179
|
Venugopal G, Mueller M, Hartmann S, Steinfelder S. Differential immunomodulation in human monocytes versus macrophages by filarial cystatin. PLoS One 2017; 12:e0188138. [PMID: 29141050 PMCID: PMC5687743 DOI: 10.1371/journal.pone.0188138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 11/01/2017] [Indexed: 12/19/2022] Open
Abstract
Parasitic nematodes have evolved powerful immunomodulatory molecules to enable their survival in immunocompetent hosts by subverting immune responses and minimizing pathological processes. One filarial molecule known to counteract host immune responses by inducing IL-10 and regulatory macrophages in mice is filarial cystatin. During a patent filarial infection monocytes encounter microfilariae in the blood, an event that occurs in asymptomatically infected filariasis patients that are immunologically hyporeactive. The microfilarial larval stage was formerly shown to induce human regulatory monocytes and macrophages. Thus, here we aim was to determine how filarial cystatin of the human pathogenic filaria Brugia malayi (BmCPI-2) contributes to immune hyporesponsiveness in human monocytes and macrophages elicited by microfilaria. For this purpose, filarial cystatin was depleted from microfilarial lysate (Mf). Detecting the immunomodulatory potential of cystatin-depleted Mf revealed that IL-10, but not IL-8 and IL-6 induction in monocytes and macrophages is dependent on the presence of cystatin. In addition, the Mf-induced expression of the regulatory surface markers PD-L1 and PD-L2 in human monocytes, but not in macrophages, is dependent on cystatin. While Mf-treated monocytes result in decreased CD4+ T-cell proliferation in a co-culture assay, stimulation of T-cells with human monocytes treated with cystatin-depleted Mf lead to a restoration of CD4+ T-cell proliferation. Moreover, IL-10 induction by cystatin within Mf was dependent on p38 and ERK in macrophages, but independent of the ERK pathway in monocytes. These findings indicate that filarial nematodes differentially trigger and exploit various signaling pathways to induce immunomodulation in different myeloid cell subsets.
Collapse
Affiliation(s)
- Gopinath Venugopal
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Marion Mueller
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Susanne Hartmann
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
- * E-mail:
| | - Svenja Steinfelder
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
180
|
Peat FJ, Colbath AC, Bentsen LM, Goodrich LR, King MR. In Vitro Effects of High-Intensity Laser Photobiomodulation on Equine Bone Marrow-Derived Mesenchymal Stem Cell Viability and Cytokine Expression. Photomed Laser Surg 2017; 36:83-91. [PMID: 29131717 DOI: 10.1089/pho.2017.4344] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE This study aimed to examine the influence of neodymium-doped yttrium aluminum garnet (Nd:YAG) laser irradiation on equine bone marrow-derived mesenchymal stem cell (MSC) viability, proliferation, and cytokine expression in vitro. BACKGROUND Photobiomodulation of cells using monochromatic light is a technique designed to influence cellular processes. Previous studies have shown dose-dependent effects of low-level laser irradiation on cell proliferation and cytokine expression in a range of cell types and species. Evidence for the influence of 1064 nm wavelength near-infrared irradiation on MSCs is sparse, and high-energy doses have shown inhibitory effects. METHODS MSC cultures from six horses were exposed to 1064 nm irradiation with an energy density of 9.77 J/cm2 and a mean output power of 13.0 W for 10 sec. MSC viability and proliferation were evaluated through flow cytometry and real-time live cell analysis. Gene expression and cytokine production in the first 24 h after irradiation were analyzed through polymerase chain reaction (PCR), multiplex assay, and enzyme-linked immunosorbent assay. RESULTS No difference in viability was detected between irradiated and control MSCs. Irradiated cells demonstrated slightly lower proliferation rates, but remained within 3.5% confluence of control cells. Twenty-four hours after irradiation, irradiated MSCs demonstrated a significant increase in expression of interleukin (IL)-10 and vascular endothelial growth factor (VEGF) compared with control MSCs. CONCLUSIONS Under these irradiation parameters, equine MSCs remained viable and expressed increased concentrations of IL-10 and VEGF. IL-10 has an anti-inflammatory action by inhibiting the synthesis of proinflammatory cytokines at the transcriptional level. This response to 1064 nm irradiation shows promise in the photobiomodulation of MSCs to enhance their therapeutic properties.
Collapse
Affiliation(s)
- Frances J Peat
- Orthopaedic Research Center, Colorado State University Veterinary Teaching Hospital , Fort Collins, Colorado
| | - Aimee C Colbath
- Orthopaedic Research Center, Colorado State University Veterinary Teaching Hospital , Fort Collins, Colorado
| | - Lori M Bentsen
- Orthopaedic Research Center, Colorado State University Veterinary Teaching Hospital , Fort Collins, Colorado
| | - Laurie R Goodrich
- Orthopaedic Research Center, Colorado State University Veterinary Teaching Hospital , Fort Collins, Colorado
| | - Melissa R King
- Orthopaedic Research Center, Colorado State University Veterinary Teaching Hospital , Fort Collins, Colorado
| |
Collapse
|
181
|
Elsegood CL, Tirnitz-Parker JE, Olynyk JK, Yeoh GC. Immune checkpoint inhibition: prospects for prevention and therapy of hepatocellular carcinoma. Clin Transl Immunology 2017; 6:e161. [PMID: 29326816 PMCID: PMC5704099 DOI: 10.1038/cti.2017.47] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 08/10/2017] [Accepted: 09/10/2017] [Indexed: 02/06/2023] Open
Abstract
The global prevalence of liver cancer is rapidly rising, mostly as a result of the amplified incidence rates of viral hepatitis, alcohol abuse and obesity in recent decades. Treatment options for liver cancer are remarkably limited with sorafenib being the gold standard for advanced, unresectable hepatocellular carcinoma but offering extremely limited improvement of survival time. The immune system is now recognised as a key regulator of cancer development through its ability to protect against infection and chronic inflammation, which promote cancer development, and eliminate tumour cells when present. However, the tolerogenic nature of the liver means that the immune response to infection, chronic inflammation and tumour cells within the hepatic environment is usually ineffective. Here we review the roles that immune cells and cytokines have in the development of the most common primary liver cancer, hepatocellular carcinoma (HCC). We then examine how the immune system may be subverted throughout the stages of HCC development, particularly with respect to immune inhibitory molecules, also known as immune checkpoints, such as programmed cell death protein-1, programmed cell death 1 ligand 1 and cytotoxic T lymphocyte antigen 4, which have become therapeutic targets. Finally, we assess preclinical and clinical studies where immune checkpoint inhibitors have been used to modify disease during the carcinogenic process. In conclusion, inhibitory molecule-based immunotherapy for HCC is in its infancy and further detailed research in relevant in vivo models is required before its full potential can be realised.
Collapse
Affiliation(s)
- Caryn L Elsegood
- School of Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Janina Ee Tirnitz-Parker
- School of Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - John K Olynyk
- School of Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,Department of Gastroenterology and Hepatology, Fiona Stanley and Fremantle Hospitals, South Metropolitan Health Service, Murdoch, Western Australia, Australia.,School of Health and Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - George Ct Yeoh
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
182
|
Traupe H. Psoriasis and the interleukin-10 family: evidence for a protective genetic effect, but not an easy target as a drug. Br J Dermatol 2017; 176:1438-1439. [PMID: 28581220 DOI: 10.1111/bjd.15158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- H Traupe
- Department of Dermatology, University Hospital of Münster, Von-Esmarch-Str. 58, 48149, Münster, Germany
| |
Collapse
|
183
|
Khezri S, Abtahi Froushani SM, Shahmoradi M. Nicotine Augments the Beneficial Effects of Mesenchymal Stem Cell-based Therapy in Rat Model of Multiple Sclerosis. Immunol Invest 2017; 47:113-124. [DOI: 10.1080/08820139.2017.1391841] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Shiva Khezri
- Department of biology, Faculty of science, Urmia University, Urmia, Iran
| | | | - Mozhgan Shahmoradi
- Department of biology, Faculty of science, Urmia University, Urmia, Iran
| |
Collapse
|
184
|
Wilbers RHP, van Raaij DR, Westerhof LB, Bakker J, Smant G, Schots A. Re-evaluation of IL-10 signaling reveals novel insights on the contribution of the intracellular domain of the IL-10R2 chain. PLoS One 2017; 12:e0186317. [PMID: 29016674 PMCID: PMC5634637 DOI: 10.1371/journal.pone.0186317] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/28/2017] [Indexed: 01/25/2023] Open
Abstract
Interleukin-10 (IL-10) is an anti-inflammatory cytokine that plays a key role in maintaining immune homeostasis. IL-10-mediated responses are triggered upon binding to a heterodimeric receptor complex consisting of IL-10 receptor (IL-10R)1 and IL-10R2. Engagement of the IL-10R complex activates the intracellular kinases Jak1 and Tyk2, but the exact roles of IL-10R2 and IL-10R2-associated signaling via Tyk2 remain unclear. To elucidate the contribution of IL-10R2 and its signaling to IL-10 activity, we re-evaluated IL-10-mediated responses on bone marrow-derived dendritic cells, macrophages and mast cells. By using bone marrow from IL-10R-/- mice it was revealed that IL-10-mediated responses depend on both IL-10R1 and IL-10R2 in all three cell types. On the contrary, bone marrow-derived cells from Tyk2-/- mice showed similar responses to IL-10 as wild-type cells, indicating that signaling via this IL-10R2-associated kinase only plays a limited role. Tyk2 was shown to control the amplitude of STAT3 activation and the up-regulation of downstream SOCS3 expression. SOCS3 up-regulation was found to be cell-type dependent and correlated with the lack of early suppression of LPS-induced TNF-α in dendritic cells. Further investigation of the IL-10R complex revealed that both the extracellular and intracellular domains of IL-10R2 influence the conformation of IL-10R1 and that both domains were required for transducing IL-10 signals. This observation highlights a novel role for the intracellular domain of IL-10R2 in the molecular mechanisms of IL-10R activation.
Collapse
Affiliation(s)
- Ruud H. P. Wilbers
- Wageningen University and Research, Plant Sciences Group, Laboratory of Nematology, Wageningen, The Netherlands
| | - Debbie R. van Raaij
- Wageningen University and Research, Plant Sciences Group, Laboratory of Nematology, Wageningen, The Netherlands
| | - Lotte B. Westerhof
- Wageningen University and Research, Plant Sciences Group, Laboratory of Nematology, Wageningen, The Netherlands
| | - Jaap Bakker
- Wageningen University and Research, Plant Sciences Group, Laboratory of Nematology, Wageningen, The Netherlands
| | - Geert Smant
- Wageningen University and Research, Plant Sciences Group, Laboratory of Nematology, Wageningen, The Netherlands
| | - Arjen Schots
- Wageningen University and Research, Plant Sciences Group, Laboratory of Nematology, Wageningen, The Netherlands
| |
Collapse
|
185
|
Cambier L, de Couto G, Ibrahim A, Echavez AK, Valle J, Liu W, Kreke M, Smith RR, Marbán L, Marbán E. Y RNA fragment in extracellular vesicles confers cardioprotection via modulation of IL-10 expression and secretion. EMBO Mol Med 2017; 9:337-352. [PMID: 28167565 PMCID: PMC5331234 DOI: 10.15252/emmm.201606924] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cardiosphere‐derived cells (CDCs) reduce myocardial infarct size via secreted extracellular vesicles (CDC‐EVs), including exosomes, which alter macrophage polarization. We questioned whether short non‐coding RNA species of unknown function within CDC‐EVs contribute to cardioprotection. The most abundant RNA species in CDC‐EVs is a Y RNA fragment (EV‐YF1); its relative abundance in CDC‐EVs correlates with CDC potency in vivo. Fluorescently labeled EV‐YF1 is actively transferred from CDCs to target macrophages via CDC‐EVs. Direct transfection of macrophages with EV‐YF1 induced transcription and secretion of IL‐10. When cocultured with rat cardiomyocytes, EV‐YF1‐primed macrophages were potently cytoprotective toward oxidatively stressed cardiomyocytes through induction of IL‐10. In vivo, intracoronary injection of EV‐YF1 following ischemia/reperfusion reduced infarct size. A fragment of Y RNA, highly enriched in CDC‐EVs, alters Il10 gene expression and enhances IL‐10 protein secretion. The demonstration that EV‐YF1 confers cardioprotection highlights the potential importance of diverse exosomal contents of unknown function, above and beyond the usual suspects (e.g., microRNAs and proteins).
Collapse
Affiliation(s)
- Linda Cambier
- Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Geoffrey de Couto
- Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Antonio K Echavez
- Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jackelyn Valle
- Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Weixin Liu
- Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | | | - Eduardo Marbán
- Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
186
|
Varzandian B, Ghaderi-Zefrehei M, Hosseinzadeh S, Sayyadi M, Taghadosi V, Varzandian S. An Investigation on the Expression Level of Interleukin 10 (IL-10) in the Healthy and Mastitic Holstein Cows and the Bioinformatics Analysis of Nucleosome Profile. Anim Biotechnol 2017; 28:294-300. [PMID: 28267404 DOI: 10.1080/10495398.2017.1283322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cytokines are immune regulators that play an essential role in regulating immune response against various infections. The present study focused on the possible association between the expression level of Interleukin 10 (IL-10) in blood and milk samples of 25 healthy and 25 mastitic cows in Fars province, Iran, using a quantitative real-time PCR assay. The experimental groups were categorized according to the number of calvings. The expression level of IL-10 was significantly higher in the blood and milk samples of mastitic cows compared to the healthy ones. Concomitant to increasing the number of calving, a numerical elevation in the expression of IL-10 in blood was observed (P < 0.05). The bioinformatics analysis of IL-10 gene revealed the promoter, exon-intron regions, and nucleosome profile. The nucleosome occupancy site was finally predicted using NUPOP software. Our result indicated that the promoter was not exactly placed in the nucleosome region, which was finally aimed to predict the position and expression of IL-10 gene in the mastitic cows.
Collapse
Affiliation(s)
- Bahareh Varzandian
- a Department of Animal Breeding, School of Agriculture , Yasouj University , Yasouj , Iran
| | | | - Saeid Hosseinzadeh
- b Department of Food Hygiene and Public Health, School of Veterinary Medicine , Shiraz University , Shiraz , Iran
| | - Mostafa Sayyadi
- b Department of Food Hygiene and Public Health, School of Veterinary Medicine , Shiraz University , Shiraz , Iran
| | - Vahideh Taghadosi
- b Department of Food Hygiene and Public Health, School of Veterinary Medicine , Shiraz University , Shiraz , Iran
| | - Sara Varzandian
- c Department of Clinical Sciences, School of Veterinary Medicine, Kazerun Branch , Islamic Azad University , Kazerun , Iran
| |
Collapse
|
187
|
Cho Y, Song MK, Kim TS, Ryu JC. Identification of novel cytokine biomarkers of hexanal exposure associated with pulmonary toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 229:810-817. [PMID: 28779894 DOI: 10.1016/j.envpol.2017.06.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 06/07/2023]
Abstract
We aimed to investigate whether exposure to low-molecular-weight saturated aliphatic aldehydes induces an airway inflammation related to lung toxicity. In previous studies, we identified that several aldehydes induced inflammatory responses through the secretion of pro-inflammatory cytokines. Here, we elucidate on whether hexanal exposure induces the lung inflammatory response through the secretion of cytokines. Hexanal is one of the aldehydes, which are major components of indoor environmental irritants. Based on a multiplexed cytokine antibody array, we investigated the cytokine expression profiles to identify the significant biomarkers of hexanal exposure and to predict the possibility of adverse effects on pulmonary toxicity using in vitro and in vivo model systems. We identified the cytokines as biomarkers involved in LEPTIN, Interleukin(IL)-10, MCP-1, and VEGF that showed similar expression patterns in both in vitro and in vivo models under hexanal exposure. These cytokines are known to be associated with diverse lung diseases, such as lung fibrosis, chronic obstructive pulmonary disease (COPD), and non-small cell lung cancer. Although further studies are needed to identify the mechanisms that underlie hexanal pulmonary toxicity, these results provide the key cytokine biomarkers in response to hexanal exposure and indicate meaningful mechanistic previewing that can be indirectly attributed to lung disease.
Collapse
Affiliation(s)
- Yoon Cho
- Center for Environment, Health and Welfare Research, Cellular and Molecular Toxicology Laboratory, Korea Institute of Science and Technology (KIST), Republic of Korea; Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Republic of Korea
| | - Mi-Kyung Song
- National Center for Efficacy Evaluation for Respiratory Disease Product, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, Republic of Korea
| | - Tae Sung Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Republic of Korea
| | - Jae-Chun Ryu
- Center for Environment, Health and Welfare Research, Cellular and Molecular Toxicology Laboratory, Korea Institute of Science and Technology (KIST), Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Republic of Korea.
| |
Collapse
|
188
|
|
189
|
Kim H, Kim YJ, Jeong HY, Kim JY, Choi EK, Chae SW, Kwon O. A standardized extract of the fruit of Hovenia dulcis alleviated alcohol-induced hangover in healthy subjects with heterozygous ALDH2: A randomized, controlled, crossover trial. JOURNAL OF ETHNOPHARMACOLOGY 2017; 209:167-174. [PMID: 28750942 DOI: 10.1016/j.jep.2017.07.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 07/06/2017] [Accepted: 07/17/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hovenia dulcis, known as the oriental raisin tree, is mainly found in East Asia. It has long been used as traditional folk remedies for alcohol intoxication. AIM OF THE STUDY To examine the anti-hangover effect of Hovenia dulcis Thunb. fruit extract (HDE) in a randomized controlled crossover trial. MATERIALS AND METHODS Twenty-six eligible male adults with heterozygous ALDH2 (23.7±0.3 years old) consumed 360mL of Korean Soju (50g alcohol) together with HDE (2460mg) or matched placebo with subsequent crossover. The blood samples were taken at baseline and 1, 4, and 12h post-treatment. RESULTS Blood alcohol, acetaldehyde, and total hangover scores were highest at 1h post-treatment with no difference between groups, but declines in hangover symptom scores were significant in the HDE group compared to the placebo group. Significant differences between groups were also observed on interleukin (IL)-6, IL-10, IL-10/IL-6 ratio, and aspartate aminotransferase levels, but not on endotoxins. Pearson correlation analysis revealed a positive correlation between total hangover symptom scores and IL-6 and IL-10 level. Further analyses by CYP2E1 polymorphism at rs10776687, rs2031920, rs3813867, and rs4838767 alleles showed a reversed association, suggesting that CYP2E1 polymorphism might be an effect modifier. CONCLUSIONS The results suggest that a favorable effect of HDE on alcohol hangovers might be associated with enhancing homeostatic regulation of inflammatory response. The magnitude of impact might be different in the presence of CYP2E1 polymorphism.
Collapse
Affiliation(s)
- Hoejin Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - You Jin Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Hye Yun Jeong
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Ji Yeon Kim
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Eun-Kyung Choi
- Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Soo Wan Chae
- Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Republic of Korea.
| | - Oran Kwon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
190
|
Naing A, Papadopoulos KP, Autio KA, Ott PA, Patel MR, Wong DJ, Falchook GS, Pant S, Whiteside M, Rasco DR, Mumm JB, Chan IH, Bendell JC, Bauer TM, Colen RR, Hong DS, Van Vlasselaer P, Tannir NM, Oft M, Infante JR. Safety, Antitumor Activity, and Immune Activation of Pegylated Recombinant Human Interleukin-10 (AM0010) in Patients With Advanced Solid Tumors. J Clin Oncol 2017; 34:3562-3569. [PMID: 27528724 DOI: 10.1200/jco.2016.68.1106] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Purpose Interleukin-10 (IL-10) stimulates the expansion and cytotoxicity of tumor-infiltrating CD8+ T cells and inhibits inflammatory CD4+ T cells. Pegylation prolongs the serum concentration of IL-10 without changing the immunologic profile. This phase I study sought to determine the safety and antitumor activity of AM0010. Patients and Methods Patients with selected advanced solid tumors were treated with AM0010 in a dose-escalation study, which was followed by a renal cell cancer (RCC) dose-expansion cohort. AM0010 was self-administered subcutaneously at doses of 1 to 40 μg/kg once per day. Primary end points were safety and tolerability; clinical activity and immune activation were secondary end points. Results In the dose-escalation and -expansion cohorts, 33 and 18 patients, respectively, were treated with daily subcutaneous injection of AM0010. AM0010 was tolerated in a heavily pretreated patient population. Treatment-related adverse events (AEs) included anemia, fatigue, thrombocytopenia, fever, and injection site reactions. Grade 3 to 4 nonhematopoietic treatment-related AEs, including rash (n = 2) and transaminitis (n = 1), were observed in five of 33 patients. Grade 3 to 4 anemia or thrombocytopenia was observed in five patients. Most treatment-related AEs were transient or reversible. AM0010 led to systemic immune activation with elevated immune-stimulatory cytokines and reduced transforming growth factor beta in the serum. Partial responses were observed in one patient with uveal melanoma and four of 15 evaluable patients with RCC treated at 20 μg/kg (overall response rate, 27%). Prolonged stable disease of at least 4 months was observed in four patients, including one with colorectal cancer with disease stabilization for 20 months. Conclusion AM0010 has an acceptable toxicity profile with early evidence of antitumor activity, particularly in RCC. These data support the further evaluation of AM0010 both alone and in combination with other immune therapies and chemotherapies.
Collapse
Affiliation(s)
- Aung Naing
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Kyriakos P Papadopoulos
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Karen A Autio
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Patrick A Ott
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Manish R Patel
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Deborah J Wong
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Gerald S Falchook
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Shubham Pant
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Melinda Whiteside
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Drew R Rasco
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - John B Mumm
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Ivan H Chan
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Johanna C Bendell
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Todd M Bauer
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Rivka R Colen
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - David S Hong
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Peter Van Vlasselaer
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Nizar M Tannir
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Martin Oft
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Jeffrey R Infante
- Aung Naing, Rivka R. Colen, David S. Hong, and Nizar M. Tannir, MD Anderson Cancer Center, Houston; Kyriakos P. Papadopoulos and Drew R. Rasco, START Center for Cancer Care, San Antonio, TX; Karen A. Autio, Memorial Sloan Kettering Cancer Center, New York, NY; Patrick A. Ott, Dana-Farber Cancer Institute, Boston, MA; Manish R. Patel, Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL; Deborah J. Wong, University of California Los Angeles, Los Angeles; Melinda Whiteside, John B. Mumm, Ivan H. Chan, Peter Van Vlasselaer, and Martin Oft, ARMO BioSciences, Redwood City, CA; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Shubham Pant, Oklahoma University, Oklahoma City, OK; and Johanna C. Bendell, Todd M. Bauer, and Jeffrey R. Infante, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| |
Collapse
|
191
|
Dietary onion ameliorates antioxidant defence, inflammatory response, and cardiovascular risk biomarkers in hypercholesterolemic Wistar rats. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
192
|
Nunez Lopez O, Cambiaso-Daniel J, Branski LK, Norbury WB, Herndon DN. Predicting and managing sepsis in burn patients: current perspectives. Ther Clin Risk Manag 2017; 13:1107-1117. [PMID: 28894374 PMCID: PMC5584891 DOI: 10.2147/tcrm.s119938] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Modern burn care has led to unprecedented survival rates in burn patients whose injuries were fatal a few decades ago. Along with improved survival, new challenges have emerged in the management of burn patients. Infections top the list of the most common complication after burns, and sepsis is the leading cause of death in both adult and pediatric burn patients. The diagnosis and management of sepsis in burns is complex as a tremendous hypermetabolic response secondary to burn injury can be superimposed on systemic infection, leading to organ dysfunction. The management of a septic burn patient represents a challenging scenario that is commonly encountered by providers caring for burn patients despite preventive efforts. Here, we discuss the current perspectives in the diagnosis and treatment of sepsis and septic shock in burn patients.
Collapse
Affiliation(s)
- Omar Nunez Lopez
- Department of Surgery, University of Texas Medical Branch.,Shriners Hospitals for Children, Galveston, TX, USA
| | - Janos Cambiaso-Daniel
- Department of Surgery, University of Texas Medical Branch.,Shriners Hospitals for Children, Galveston, TX, USA.,Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Ludwik K Branski
- Department of Surgery, University of Texas Medical Branch.,Shriners Hospitals for Children, Galveston, TX, USA
| | - William B Norbury
- Department of Surgery, University of Texas Medical Branch.,Shriners Hospitals for Children, Galveston, TX, USA
| | - David N Herndon
- Department of Surgery, University of Texas Medical Branch.,Shriners Hospitals for Children, Galveston, TX, USA.,Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
193
|
Singh P, Castillo A, Islam MT, Majid DSA. Evidence for Prohypertensive, Proinflammatory Effect of Interleukin-10 During Chronic High Salt Intake in the Condition of Elevated Angiotensin II Level. Hypertension 2017; 70:839-845. [PMID: 28847894 DOI: 10.1161/hypertensionaha.117.09401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 03/30/2017] [Accepted: 07/07/2017] [Indexed: 12/31/2022]
Abstract
IL-10 (interleukin-10) has been suggested to play a protective role in angiotensin II (AngII)-induced cardiovascular disorders. This study examined the role of endogenous IL-10 in salt-sensitive hypertension and renal injury induced by AngII. Responses to chronic AngII (400 ng/min per kilogram body weight; osmotic minipump) infusion were evaluated in IL-10 gene knockout mice fed with either normal salt diet (0.3% NaCl) or high salt (HS; 4% NaCl) diet, and these responses were compared with those in wild-type mice. Normal salt diets or HS diets were given alone for the first 2 weeks and then with AngII treatment for an additional 2 weeks (n=6 in each group). Arterial pressure was continuously monitored by implanted radio-telemetry, and a 24-hour urine collection was performed by metabolic cages on the last day of the experimental period. Basal mean arterial pressure was lower in IL-10 gene knockout mice than in wild-type (98±3 versus 113±3 mm Hg) mice. Mean arterial pressure responses to normal salt/HS alone or to the AngII+normal salt treatment were similar in both strains. However, the increase in mean arterial pressure induced by the AngII+HS treatment was significantly lower in IL-10 gene knockout mice (15±5% versus 37±3%) compared with wild-type mice. Renal tissue endothelial nitric oxide synthase expression (≈3-folds) and urinary excretion of nitric oxide metabolites, nitrate/nitrite (1.2±0.1 versus 0.2±0.02 µmol/L/24 hours) were higher in IL-10 gene knockout mice compared with wild-type mice. These results indicate that an increase in nitric oxide production helps to mitigate salt-sensitive hypertension induced by AngII and suggest that a compensatory interaction between IL-10 and nitric oxide exists in modulating AngII-induced responses during HS intake.
Collapse
Affiliation(s)
- Purnima Singh
- From the Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Alexander Castillo
- From the Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - M Toriqul Islam
- From the Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Dewan S A Majid
- From the Department of Physiology, Tulane University School of Medicine, New Orleans, LA.
| |
Collapse
|
194
|
Hosseini A, Estiri H, Akhavan Niaki H, Alizadeh A, Abdolhossein Zadeh B, Ghaderian SMH, Farjadfar A, Fallah A. Multiple Sclerosis Gene Therapy with Recombinant Viral Vectors: Overexpression of IL-4, Leukemia Inhibitory Factor, and IL-10 in Wharton's Jelly Stem Cells Used in EAE Mice Model. CELL JOURNAL 2017; 19:361-374. [PMID: 28836399 PMCID: PMC5570402 DOI: 10.22074/cellj.2017.4497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 09/07/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Immunotherapy and gene therapy play important roles in modern medicine. The aim of this study is to evaluate the overexpression of interleukin-4 (IL-4), IL-10 and leukemia inhibitory factor (LIF) in Wharton's jelly stem cells (WJSCs) in the experimental autoimmune encephalomyelitis (EAE) mice model. MATERIALS AND METHODS In this experimental study, a DNA construction containing IL- 4, IL-10 and LIF was assembled to make a polycistronic vector (as the transfer vector). Transfer and control vectors were co-transfected into Human Embryonic Kidney 293 (HEK-293T) cells with helper plasmids which produced recombinant lentiviral viruses (rLV). WJSCs were transduced with rLV to make recombinant WJSC (rWJSC). In vitro protein and mRNA overexpression of IL-4, LIF, and IL-10 were evaluated using quantitative polymerase chain reaction (qPCR), enzyme-linked immunosorbent assay (ELISA) and western blot (WB) analysis. EAE was induced in mice by MOG-CFA and pertussis toxin. EAE mice were injected twice with 2×105 rWJSCs. The in vivo level of IL-4, LIF, IL-10 cytokines and IL-17 were measured by ELISA. Brain tissues were analyzed histologically for evaluation of EAE lesions. RESULTS Isolated WJSCs were performed to characterize by in vitro differentiation and surface markers were analyzed by flow cytometry method. Cloning of a single lentiviral vector with five genes was done successfully. Transfection of transfer and control vectors were processed based on CaPO4 method with >90% efficiency. Recombinant viruses were produced and results of titration showed 2-3×107 infection-unit/ml. WJSCs were transduced using recombinant viruses. IL-4, IL-10 and LIF overexpression were confirmed by ELISA, WB and qPCR. The EAE mice treated with rWJSC showed reduction of Il-17, and brain lesions as well as brain cellular infiltration, in vivo. Weights and physical activity were improved in gene-treated group. CONCLUSIONS These results showed that gene therapy using anti-inflammatory cytokines can be a promising approach against multiple sclerosis (MS). In addition, considering the immunomodulatory potential of WJSCs, an approach using a combination of WJSCs and gene therapy will enhance the treatment efficacy.
Collapse
Affiliation(s)
- Ahmad Hosseini
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Cell Biology and Anatomical Science, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hajar Estiri
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Iranian Institute of Cell and Gene Therapy, Tehran, Iran
| | - Haleh Akhavan Niaki
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Babol, Iran.,Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Akram Alizadeh
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Baharak Abdolhossein Zadeh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Akbar Farjadfar
- Department of Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Fallah
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,BioViva USA Inc, Bainbridge Island WA, USA
| |
Collapse
|
195
|
Johannessen L, Sundberg TB, O'Connell DJ, Kolde R, Berstler J, Billings KJ, Khor B, Seashore-Ludlow B, Fassl A, Russell CN, Latorre IJ, Jiang B, Graham DB, Perez JR, Sicinski P, Phillips AJ, Schreiber SL, Gray NS, Shamji AF, Xavier RJ. Small-molecule studies identify CDK8 as a regulator of IL-10 in myeloid cells. Nat Chem Biol 2017; 13:1102-1108. [PMID: 28805801 DOI: 10.1038/nchembio.2458] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/17/2017] [Indexed: 12/27/2022]
Abstract
Enhancing production of the anti-inflammatory cytokine interleukin-10 (IL-10) is a promising strategy to suppress pathogenic inflammation. To identify new mechanisms regulating IL-10 production, we conducted a phenotypic screen for small molecules that enhance IL-10 secretion from activated dendritic cells. Mechanism-of-action studies using a prioritized hit from the screen, BRD6989, identified the Mediator-associated kinase CDK8, and its paralog CDK19, as negative regulators of IL-10 production during innate immune activation. The ability of BRD6989 to upregulate IL-10 is recapitulated by multiple, structurally differentiated CDK8 and CDK19 inhibitors and requires an intact cyclin C-CDK8 complex. Using a highly parallel pathway reporter assay, we identified a role for enhanced AP-1 activity in IL-10 potentiation following CDK8 and CDK19 inhibition, an effect associated with reduced phosphorylation of a negative regulatory site on c-Jun. These findings identify a function for CDK8 and CDK19 in regulating innate immune activation and suggest that these kinases may warrant consideration as therapeutic targets for inflammatory disorders.
Collapse
Affiliation(s)
- Liv Johannessen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Thomas B Sundberg
- Center for the Development of Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
| | - Daniel J O'Connell
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Raivo Kolde
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James Berstler
- Center for the Development of Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
| | - Katelyn J Billings
- Department of Chemistry, Yale University, New Haven, Connecticut, USA.,Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
| | - Bernard Khor
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlin N Russell
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Isabel J Latorre
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Baishan Jiang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Daniel B Graham
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, Massachusetts, USA.,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jose R Perez
- Center for the Development of Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew J Phillips
- Center for the Development of Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
| | - Stuart L Schreiber
- Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts, USA.,Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Cambridge, Massachusetts, USA
| | - Nathanael S Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Alykhan F Shamji
- Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
| | - Ramnik J Xavier
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, Massachusetts, USA.,Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
196
|
Antioxidant and Anti-Inflammatory Effects of Shungite against Ultraviolet B Irradiation-Induced Skin Damage in Hairless Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7340143. [PMID: 28894510 PMCID: PMC5574306 DOI: 10.1155/2017/7340143] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/14/2017] [Accepted: 07/04/2017] [Indexed: 12/22/2022]
Abstract
As fullerene-based compound applications have been rapidly increasing in the health industry, the need of biomedical research is urgently in demand. While shungite is regarded as a natural source of fullerene, it remains poorly documented. Here, we explored the in vivo effects of shungite against ultraviolet B- (UVB-) induced skin damage by investigating the physiological skin parameters, immune-redox profiling, and oxidative stress molecular signaling. Toward this, mice were UVB-irradiated with 0.75 mW/cm2 for two consecutive days. Consecutively, shungite was topically applied on the dorsal side of the mice for 7 days. First, we found significant improvements in the skin parameters of the shungite-treated groups revealed by the reduction in roughness, pigmentation, and wrinkle measurement. Second, the immunokine profiling in mouse serum and skin lysates showed a reduction in the proinflammatory response in the shungite-treated groups. Accordingly, the redox profile of shungite-treated groups showed counterbalance of ROS/RNS and superoxide levels in serum and skin lysates. Last, we have confirmed the involvement of Nrf2- and MAPK-mediated oxidative stress pathways in the antioxidant mechanism of shungite. Collectively, the results clearly show that shungite has an antioxidant and anti-inflammatory action against UVB-induced skin damage in hairless mice.
Collapse
|
197
|
High-density lipoprotein immunomodulates the functional activities of macrophage and cytokines produced during ex vivo macrophage-CD4 + T cell crosstalk at the recent-onset human type 1 diabetes. Cytokine 2017; 96:59-70. [DOI: 10.1016/j.cyto.2017.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/08/2017] [Accepted: 03/01/2017] [Indexed: 12/31/2022]
|
198
|
The Influence and Delivery of Cytokines and their Mediating Effect on Muscle Satellite Cells. CURRENT STEM CELL REPORTS 2017. [DOI: 10.1007/s40778-017-0089-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
199
|
Nilsson T, Gielis JF, Slama A, Hansson C, Wallinder A, Ricksten SE, Dellgren G. Comparison of two strategies for ex vivo lung perfusion. J Heart Lung Transplant 2017; 37:S1053-2498(17)31883-1. [PMID: 28756120 DOI: 10.1016/j.healun.2017.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/03/2017] [Accepted: 07/03/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Two clinically used strategies for ex vivo lung perfusion (EVLP) were compared in a porcine model with respect to lung function, metabolism, inflammatory response, oxidative stress, and cell viability. METHODS Porcine lungs (n = 20) were preserved, harvested, and kept cooled for 2 hours. After randomization, EVLP was performed using a cellular perfusate and open left atrium (COA group) or an acellular perfusate and a closed left atrium (ACA group). Oxygenation (partial pressure of arterial oxygen/fraction of inspired oxygen), compliance, dead space, weight, and perfusate oncotic pressure were registered before and after a 4-hour period of reconditioning. Lung tissue samples were collected before and after EVLP for quantitative polymerase chain reaction analysis of gene expression for inflammatory markers, measurement of tissue hypoxia (hypoxia inducible factor-1α) and oxidative stress (ascorbyl radical), and viability (trypan blue staining) and lung histopathology. RESULTS In 3 of 10 lungs undergoing EVLP in the ACA group, EVLP was terminated prematurely because of severe lung edema and inability to perfuse the lungs. There were no significant differences in changes of lung oxygenation or pulmonary vascular resistance between groups. Compliance decreased and lung weights increased in both groups, but more in the ACA group (p = 0.083 and p = 0.065, respectively). There was no obvious difference in gene expression for hypoxia inducible factor-1α, inflammatory markers, free radicals, or lung injury between groups. CONCLUSIONS Lung edema formation and decreased lung compliance occurs with both EVLP techniques but were more pronounced in the ACA group. Otherwise, there were no differences in lung function, inflammatory response, ischemia/reperfusion injury, or histopathologic changes between the EVLP techniques.
Collapse
Affiliation(s)
- Tobias Nilsson
- Department of Cardiothoracic Anesthesia and Intensive Care, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Anesthesiology and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan F Gielis
- Laboratory for Microbiology, Parasitology and Hygiene, Antwerp University, Antwerp, Belgium
| | - Alexis Slama
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria; Department of Thoracic Surgery and Surgical Endoscopy, Ruhrlandklinik, University Clinic Essen, Essen, Germany
| | - Christoffer Hansson
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andreas Wallinder
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sven-Erik Ricksten
- Department of Cardiothoracic Anesthesia and Intensive Care, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Anesthesiology and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Göran Dellgren
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
200
|
Cardioprotective time-window of Penehyclidine hydrochloride postconditioning: A rat study. Eur J Pharmacol 2017; 812:48-56. [PMID: 28684235 DOI: 10.1016/j.ejphar.2017.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/11/2017] [Accepted: 07/03/2017] [Indexed: 12/16/2022]
Abstract
Pharmacological postconditioning offers a clinical perspective for all patients with ischemic heart disease. Penehyclidine hydrochloride (PHC) is a new type of anticholinergic drug. We previously reported that PHC preconditioning protects against I/R injury in rat hearts in vivo. Ischemic heart disease often occurs suddenly, so postconditioning is more significant than preconditioning. However, studies evaluating myocardial protective effects of PHC postconditioning are unavailable. We explored the effects and time-window of cardioprotection of PHC postconditioning in myocardial I/R injury. PHC was administered by intravenous at various times (t = -5, 0, 5, 10, 15, or 30min) after the onset of reperfusion in addition to I/R rat. We observed five different indicators including infarct size, inflammatory response, myocardial enzyme, oxidative stress, and Ca2+ overload to quantify the effect of cardioprotection. Evans blue and TTC staining were used to measure myocardial infarct size. The expression of NF-κ B and IκB-α was analyzed using Western blot. ELISA was conducted to detect inflammatory and anti-inflammatory mediators. The Ca2+ level was determined using assay kit. PHC postconditioning (from -5 to 10min after the onset of reperfusion) significantly reduced infarct size, downregulated NF-κ B expression, and decreased the release of inflammatory mediators, while significantly upregulating IκB-α expression and increasing the release of anti-inflammatory mediators. All PHC postconditioning groups significantly reduced Ca2+ level. PHC postconditioning is cardioprotective over a larger time-window (from -5 to 10min after the onset of reperfusion). The probable mechanism is inhibition of NF-кB regulated inflammatory response pathway.
Collapse
|