51
|
Zhao X, Dong B, Friesen M, Liu S, Zhu C, Yang C. Capsaicin Attenuates Lipopolysaccharide-Induced Inflammation and Barrier Dysfunction in Intestinal Porcine Epithelial Cell Line-J2. Front Physiol 2021; 12:715469. [PMID: 34630139 PMCID: PMC8497985 DOI: 10.3389/fphys.2021.715469] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/30/2021] [Indexed: 11/13/2022] Open
Abstract
Capsaicin is a spicy, highly pungent, colorless, vanilloid compound found in chili peppers with anti-inflammatory, antioxidant, anti-cancer, and analgesic properties. However, the protective effects of capsaicin on the pig intestine during inflammation are yet to be explored. This study investigated the effects of capsaicin on the gut inflammatory response, intestinal epithelial integrity, and gene expression level of nutrient transporters in a model of lipopolysaccharide (LPS)-induced inflammation in non-differentiated intestinal porcine epithelial cell line-J2 (IPEC-J2). The results showed that the pre-treatment of cells with capsaicin (100 μM) significantly decreased the gene expression and secretion of proinflammatory cytokines induced by LPS through Toll-like receptor 4 (TLR4)/NF-κB signaling pathway. In addition, pre-treatment of cells with capsaicin also increased both gene and protein abundance of tight junction proteins. Furthermore, pre-treatment cells with capsaicin significantly increased trans-epithelial electrical resistance (TEER) and decreased permeability of fluorescein isothiocyanate-dextran (FD4) from the apical side to the basolateral side compared with the control (P < 0.05). Additionally, pre-treatment of cells with capsaicin upregulated the mRNA abundance of nutrients transporters such as Na+/glucose cotransporter 1 (SGLT1). These results suggested that capsaicin could attenuate LPS-induced inflammation response through TLR4/NF-κB pathway and improve barrier integrity and glucose absorption.
Collapse
Affiliation(s)
- Xiaoya Zhao
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Bingqi Dong
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Marissa Friesen
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Shangxi Liu
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Changqing Zhu
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, China
| | - Chengbo Yang
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
52
|
Fragopoulou E, Argyrou C, Detopoulou M, Tsitsou S, Seremeti S, Yannakoulia M, Antonopoulou S, Kolovou G, Kalogeropoulos P. The effect of moderate wine consumption on cytokine secretion by peripheral blood mononuclear cells: A randomized clinical study in coronary heart disease patients. Cytokine 2021; 146:155629. [PMID: 34247040 DOI: 10.1016/j.cyto.2021.155629] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 12/14/2022]
Abstract
Many studies conclude that wine consumption is related to lower risk for cardiovascular diseases partially through the amelioration of inflammatory biomarkers. The aim of the present study was to examine the effects of wine consumption on the inflammatory response and to compare these effects with the consumption of similar amount of alcohol without the wine micro-constituents in cardiovascular disease patients. Therefore, a randomized, single-blind, controlled, three-arm parallel intervention study was designed. Cardiovascular disease patients were randomly assigned to one of the three groups. In Group A participants consumed no alcohol, in Group B (ethanol group) and Group C (wine group) participants consumed 27 g of alcohol per day. Biological samples were collected at the beginning, on the 4th and 8th week and several biomarkers were measured. Peripheral blood mononuclear cells that were isolated from patients were incubated under basal and inflammatory conditions for 4 and 24 h and the secretion of interleukin 1β (IL-1β) and tumor necrosis factor α (TNFα) was measured. No significant difference was observed among the three groups before the initiation or during the intervention in the most soluble biomarkers. Higher TNFα secretion by peripheral blood mononuclear cells was observed at basal conditions in the ethanol group both at 4 and 24 h of incubation versus baseline secretion. Furthermore, lower secretion of the ΤNFα was observed after 8 weeks of intake in the wine group versus the ethanol group, both at 4 and 24 h of incubation. In conclusion, the light to moderate wine consumption for 8 weeks revealed an attenuation of the ethanol consumption effect on cytokine secretion at basal conditions from the patients' peripheral blood mononuclear cells.
Collapse
Affiliation(s)
- Elizabeth Fragopoulou
- Department of Nutrition and Dietetics, Harokopio University, 70 Eleftheriou Venizelou Avenue Kallithea, 17671 Athens, Greece.
| | - Chrysa Argyrou
- Department of Nutrition and Dietetics, Harokopio University, 70 Eleftheriou Venizelou Avenue Kallithea, 17671 Athens, Greece
| | - Maria Detopoulou
- Department of Nutrition and Dietetics, Harokopio University, 70 Eleftheriou Venizelou Avenue Kallithea, 17671 Athens, Greece
| | - Sofia Tsitsou
- Department of Nutrition and Dietetics, Harokopio University, 70 Eleftheriou Venizelou Avenue Kallithea, 17671 Athens, Greece
| | - Sotiria Seremeti
- Department of Nutrition and Dietetics, Harokopio University, 70 Eleftheriou Venizelou Avenue Kallithea, 17671 Athens, Greece
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, 70 Eleftheriou Venizelou Avenue Kallithea, 17671 Athens, Greece
| | - Smaragdi Antonopoulou
- Department of Nutrition and Dietetics, Harokopio University, 70 Eleftheriou Venizelou Avenue Kallithea, 17671 Athens, Greece
| | - Genovefa Kolovou
- Cardiology Department, Onassis Cardiac Surgery Center, Athens. Greece
| | | |
Collapse
|
53
|
Kang DY, Sp N, Jo ES, Rugamba A, Kim HD, Kim IH, Park JC, Bae SW, Jang KJ, Yang YM. Non-toxic sulfur inhibits LPS-induced inflammation by regulating TLR-4 and JAK2/STAT3 through IL-6 signaling. Mol Med Rep 2021; 24:485. [PMID: 33907855 PMCID: PMC8127030 DOI: 10.3892/mmr.2021.12124] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 03/29/2021] [Indexed: 01/22/2023] Open
Abstract
Janus kinase 2 (JAK2) and STAT3 signaling is considered a major pathway in lipopolysaccharide (LPS)‑induced inflammation. Toll‑like receptor 4 (TLR‑4) is an inflammatory response receptor that activates JAK2 during inflammation. STAT3 is a transcription factor for the pro‑inflammatory cytokine IL‑6 in inflammation. Sulfur is an essential element in the amino acids and is required for growth and development. Non‑toxic sulfur (NTS) can be used in livestock feeds as it lacks toxicity. The present study aimed to inhibit LPS‑induced inflammation in C2C12 myoblasts using NTS by regulating TLR‑4 and JAK2/STAT3 signaling via the modulation of IL‑6. The 3‑(4,5‑dimethylthiazol‑2‑yl)‑2,5‑diphenyltetrazolium bromide assay was conducted to analyze cell viability and reverse transcription polymerase chain reaction and western blotting performed to measure mRNA and protein expression levels. Chromatin immunoprecipitation and enzyme‑linked immunosorbent assays were used to determine the binding activity of proteins. The results indicated that NTS demonstrated a protective effect against LPS‑induced cell death and inhibited LPS‑induced expression of TLR‑4, JAK2, STAT3 and IL‑6. In addition, NTS inhibited the expression of nuclear phosphorylated‑STAT3 and its binding to the IL‑6 promoter. Therefore, NTS may be a potential candidate drug for the treatment of inflammation.
Collapse
Affiliation(s)
- Dong Young Kang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, North Chungcheong 27478, Republic of Korea
| | - Nipin Sp
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, North Chungcheong 27478, Republic of Korea
| | - Eun Seong Jo
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, North Chungcheong 27478, Republic of Korea
| | - Alexis Rugamba
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, North Chungcheong 27478, Republic of Korea
| | - Hyoung Do Kim
- Nara Bio Co., Ltd., Gunsan, Jeollabuk-do 54006, Republic of Korea
| | - Il Ho Kim
- Nara Bio Co., Ltd., Gunsan, Jeollabuk-do 54006, Republic of Korea
| | - Jong-Chan Park
- Plant Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon 34141, Republic of Korea
| | - Se Won Bae
- Department of Chemistry and Cosmetics, Jeju National University, Jeju-si, Jeju-do 63243, Republic of Korea
| | - Kyoung-Jin Jang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, North Chungcheong 27478, Republic of Korea
| | - Young Mok Yang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, North Chungcheong 27478, Republic of Korea
| |
Collapse
|
54
|
Venusova E, Kolesarova A, Horky P, Slama P. Physiological and Immune Functions of Punicalagin. Nutrients 2021; 13:nu13072150. [PMID: 34201484 PMCID: PMC8308219 DOI: 10.3390/nu13072150] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 02/01/2023] Open
Abstract
The aim of this publication is to compile a summary of the findings regarding punicalagin in various tissues described thus far in the literature, with an emphasis on the effect of this substance on immune reactions. Punicalagin (PUN) is an ellagitannin found in the peel of pomegranate (Punica granatum). It is a polyphenol with proven antioxidant, hepatoprotective, anti-atherosclerotic and chemopreventive activities, antiproliferative activity against tumor cells; it inhibits inflammatory pathways and the action of toxic substances, and is highly tolerated. This work describes the source, metabolism, functions and effects of punicalagin, its derivatives and metabolites. Furthermore, its anti-inflammatory and antioxidant effects are described.
Collapse
Affiliation(s)
- Eva Venusova
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic;
| | - Adriana Kolesarova
- Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
| | - Pavel Horky
- Department of Animal Nutrition and Forage Production, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic;
| | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic;
- Correspondence: ; Tel.: +420-545133146
| |
Collapse
|
55
|
Moradi S, Jarrahi E, Ahmadi A, Salimian J, Karimi M, Zarei A, Azimzadeh Jamalkandi S, Ghanei M. PI3K signalling in chronic obstructive pulmonary disease and opportunities for therapy. J Pathol 2021; 254:505-518. [PMID: 33959951 DOI: 10.1002/path.5696] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/01/2021] [Accepted: 04/26/2021] [Indexed: 11/08/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease characterised by airway inflammation and progressive obstruction of the lung airflow. Current pharmacological treatments include bronchodilators, alone or in combination with steroids, or other anti-inflammatory agents, which have only partially contributed to the inhibition of disease progression and mortality. Therefore, further research unravelling the underlying mechanisms is necessary to develop new anti-COPD drugs with both lower toxicity and higher efficacy. Extrinsic signalling pathways play crucial roles in COPD development and exacerbations. In particular, phosphoinositide 3-kinase (PI3K) signalling has recently been shown to be a major driver of the COPD phenotype. Therefore, several small-molecule inhibitors have been identified to block the hyperactivation of this signalling pathway in COPD patients, many of them showing promising outcomes in both preclinical animal models of COPD and human clinical trials. In this review, we discuss the critically important roles played by hyperactivated PI3K signalling in the pathogenesis of COPD. We also critically review current therapeutics based on PI3K inhibition, and provide suggestions focusing on PI3K signalling for the further improvement of the COPD phenotype. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sharif Moradi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Esmaeil Jarrahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Karimi
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Zarei
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
56
|
Zimmerman B, Kundu P, Rooney WD, Raber J. The Effect of High Fat Diet on Cerebrovascular Health and Pathology: A Species Comparative Review. Molecules 2021; 26:3406. [PMID: 34199898 PMCID: PMC8200075 DOI: 10.3390/molecules26113406] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/19/2021] [Accepted: 06/01/2021] [Indexed: 02/08/2023] Open
Abstract
In both humans and animal models, consumption of a high-saturated-fat diet has been linked to vascular dysfunction and cognitive impairments. Laboratory animals provide excellent models for more invasive high-fat-diet-related research. However, the physiological differences between humans and common animal models in terms of how they react metabolically to high-fat diets need to be considered. Here, we review the factors that may affect the translatability of mechanistic research in animal models, paying special attention to the effects of a high-fat diet on vascular outcomes. We draw attention to the dissociation between metabolic syndrome and dyslipidemia in rodents, unlike the state in humans, where the two commonly occur. We also discuss the differential vulnerability between species to the metabolic and vascular effects of macronutrients in the diet. Findings from animal studies are better interpreted as modeling specific aspects of dysfunction. We conclude that the differences between species provide an opportunity to explore why some species are protected from the detrimental aspects of high-fat-diet-induced dysfunction, and to translate these findings into benefits for human health.
Collapse
Affiliation(s)
- Benjamin Zimmerman
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
| | - William D. Rooney
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
57
|
IL-5 mediates monocyte phenotype and pain outcomes in fibromyalgia. Pain 2021; 162:1468-1482. [PMID: 33003107 PMCID: PMC7987864 DOI: 10.1097/j.pain.0000000000002089] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/18/2020] [Indexed: 01/09/2023]
Abstract
ABSTRACT Fibromyalgia (FM) is characterized by widespread chronic pain, fatigue, and somatic symptoms. The influence of phenotypic changes in monocytes on symptoms associated with FM is not fully understood. The primary aim of this study was to take a comprehensive whole-body to molecular approach in characterizing relationships between monocyte phenotype and FM symptoms in relevant clinical populations. Lipopolysaccharide-evoked and spontaneous secretion of IL-5 and other select cytokines from circulating monocytes was higher in women with FM compared to women without pain. In addition, greater secretion of IL-5 was significantly associated with pain and other clinically relevant psychological and somatic symptoms of FM. Furthermore, higher levels of pain and pain-related symptoms were associated with a lower percentage of intermediate monocytes (CD14++/CD16+) and a greater percentage of nonclassical monocytes (CD14+/CD16++) in women with FM. Based on findings from individuals with FM, we examined the role of IL-5, an atypical cytokine secreted from monocytes, in an animal model of widespread muscle pain. Results from the animal model show that IL-5 produces analgesia and polarizes monocytes toward an anti-inflammatory phenotype (CD206+). Taken together, our data suggest that monocyte phenotype and their cytokine profiles are associated with pain-related symptoms in individuals with FM. Furthermore, our data show that IL-5 has a potential role in analgesia in an animal model of FM. Thus, targeting anti-inflammatory cytokines such as IL-5 secreted by circulating leukocytes could serve as a promising intervention to control pain and other somatic symptoms associated with FM.
Collapse
|
58
|
Guo Y, Li X, Wang Z, Yu B. Gut Microbiota Dysbiosis in Human Hypertension: A Systematic Review of Observational Studies. Front Cardiovasc Med 2021; 8:650227. [PMID: 34055933 PMCID: PMC8160125 DOI: 10.3389/fcvm.2021.650227] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/13/2021] [Indexed: 12/28/2022] Open
Abstract
Introduction: Hypertension is one of the major risk factors to human health and human studies on association between gut microbiota and hypertension or blood pressure have received increased attention. In the present study, we aim to evaluate gut microbiota dysbiosis in human hypertension using a method of systematic review. Methods: PubMed, EMBASE, and Web of Science databases were searched until March 2021 to identify eligible articles. Additional articles were also identified by searching specific authors in this field. Inclusion criteria were observational studies based on stool samples with hypertension group and control group. Newcastle-Ottawa quality assessment scale (NOS) was used to assess the quality of the included studies. PROSPERO registration number: CRD42020212219. Results: A total of 17 studies enrolling 9,085 participants were included. Fifteen of the enrolled studies showed good quality and two studies showed fair quality based on NOS. We found alpha diversity in hypertension decreased significantly and microbial structure can be separated compared with control groups. Gut microbiota of hypertension showed depletion of short chain fatty acids (SCFAs) producers and over-growth of some Proteobacteria and Bacteroidetes members. Up-regulation of lipopolysaccharide biosynthesis, phosphotransferase system, ABC transporters, etc. and down-regulation of some amino acid metabolism, etc. in hypertension were reported. Fecal SCFAs levels increased and plasma SCFAs levels decreased in hypertension. Stronger microbial interactions in hypertension were seen. Conclusion: In conclusion, gut microbiota dysbiosis was observed in hypertension, including decreased diversity, altered microbial structure, compositional change of taxa, alterations of microbial function, nutritional and immunological factors, and microbial interactions. Poor absorption and high excretion of SCFAs may play an important role in the pathogenesis of hypertension. These findings may provide insights into etiology study and new microbial-based therapies of hypertension. Systematic Review Registration: PROSPERO database, identifier CRD42020212219.
Collapse
Affiliation(s)
- Yang Guo
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xiaosu Li
- Department of Cardiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zhijian Wang
- Department of Cardiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bo Yu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| |
Collapse
|
59
|
Suttapitugsakul S, Tong M, Wu R. Time-Resolved and Comprehensive Analysis of Surface Glycoproteins Reveals Distinct Responses of Monocytes and Macrophages to Bacterial Infection. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 133:11595-11604. [PMID: 34421137 PMCID: PMC8376197 DOI: 10.1002/ange.202102692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Indexed: 12/26/2022]
Abstract
Glycoproteins on the surface of immune cells play extremely important roles in response to pathogens. Yet, a systematic and time-resolved investigation of surface glycoproteins during the immune response remains to be explored. Integrating selective enrichment of surface glycoproteins with multiplexed proteomics, we globally and site-specifically quantified the dynamics of surface glycoproteins on THP-1 monocytes and macrophages in response to bacterial infection and during the monocyte-to-macrophage differentiation. The time-resolved analysis reveals transient changes and differential remodeling of surface glycoproteins on both cell types, and potential upstream regulators and downstream effects of the regulated glycoproteins. Besides, we identified novel surface glycoproteins participating in the immune response such as APMAP, and site-specific changes of glycoproteins. This study provides unprecedented information to deepen our understanding of glycoproteins and cellular activities.
Collapse
Affiliation(s)
- Suttipong Suttapitugsakul
- School of Chemistry and Biochemistry, and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology Atlanta, GA 30332 (USA)
| | - Ming Tong
- School of Chemistry and Biochemistry, and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology Atlanta, GA 30332 (USA)
| | - Ronghu Wu
- School of Chemistry and Biochemistry, and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology Atlanta, GA 30332 (USA)
| |
Collapse
|
60
|
Sp N, Kang DY, Kim HD, Rugamba A, Jo ES, Park JC, Bae SW, Lee JM, Jang KJ. Natural Sulfurs Inhibit LPS-Induced Inflammatory Responses through NF-κB Signaling in CCD-986Sk Skin Fibroblasts. Life (Basel) 2021; 11:life11050427. [PMID: 34068523 PMCID: PMC8151259 DOI: 10.3390/life11050427] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 12/18/2022] Open
Abstract
Lipopolysaccharide (LPS)-induced inflammatory response leads to serious damage, up to and including tumorigenesis. Natural mineral sulfur, non-toxic sulfur (NTS), and methylsulfonylmethane (MSM) have anti-inflammatory activity that may inhibit LPS-induced inflammation. We hypothesized that sulfur compounds could inhibit LPS-induced inflammatory responses in CCD-986Sk skin fibroblasts. We used Western blotting and real-time PCR to analyze molecular signaling in treated and untreated cultures. We also used flow cytometry for cell surface receptor analysis, comet assays to evaluate DNA damage, and ELISA-based cytokine detection. LPS induced TLR4 activation and NF-κB signaling via canonical and protein kinase C (PKC)-dependent pathways, while NTS and MSM downregulated that response. NTS and MSM also inhibited LPS-induced nuclear accumulation and binding of NF-κB to proinflammatory cytokines COX-2, IL-1β, and IL-6. Finally, the sulfur compounds suppressed LPS-induced ROS accumulation and DNA damage in CCD-986Sk cells. These results suggest that natural sulfur compounds could be used to treat inflammation and may be useful in the development of cosmetics.
Collapse
Affiliation(s)
- Nipin Sp
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.); (H.D.K.); (A.R.)
| | - Dong Young Kang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.); (H.D.K.); (A.R.)
| | - Hyoung Do Kim
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.); (H.D.K.); (A.R.)
| | - Alexis Rugamba
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.); (H.D.K.); (A.R.)
| | - Eun Seong Jo
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Cheongju 28159, Korea; (E.S.J.); (J.-M.L.)
| | - Jong-Chan Park
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon 34141, Korea;
| | - Se Won Bae
- Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Korea;
| | - Jin-Moo Lee
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Cheongju 28159, Korea; (E.S.J.); (J.-M.L.)
| | - Kyoung-Jin Jang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.); (H.D.K.); (A.R.)
- Correspondence: ; Tel.: +82-2-2030-7812
| |
Collapse
|
61
|
Suttapitugsakul S, Tong M, Wu R. Time-Resolved and Comprehensive Analysis of Surface Glycoproteins Reveals Distinct Responses of Monocytes and Macrophages to Bacterial Infection. Angew Chem Int Ed Engl 2021; 60:11494-11503. [PMID: 33684247 PMCID: PMC8549569 DOI: 10.1002/anie.202102692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Indexed: 12/17/2022]
Abstract
Glycoproteins on the surface of immune cells play extremely important roles in response to pathogens. Yet, a systematic and time-resolved investigation of surface glycoproteins during the immune response remains to be explored. Integrating selective enrichment of surface glycoproteins with multiplexed proteomics, we globally and site-specifically quantified the dynamics of surface glycoproteins on THP-1 monocytes and macrophages in response to bacterial infection and during the monocyte-to-macrophage differentiation. The time-resolved analysis reveals transient changes and differential remodeling of surface glycoproteins on both cell types, and potential upstream regulators and downstream effects of the regulated glycoproteins. Besides, we identified novel surface glycoproteins participating in the immune response such as APMAP, and site-specific changes of glycoproteins. This study provides unprecedented information to deepen our understanding of glycoproteins and cellular activities.
Collapse
Affiliation(s)
- Suttipong Suttapitugsakul
- School of Chemistry and Biochemistry, and the Petit Institute for
Bioengineering and Bioscience, Georgia Institute of Technology Atlanta, GA 30332
(USA)
| | - Ming Tong
- School of Chemistry and Biochemistry, and the Petit Institute for
Bioengineering and Bioscience, Georgia Institute of Technology Atlanta, GA 30332
(USA)
| | - Ronghu Wu
- School of Chemistry and Biochemistry, and the Petit Institute for
Bioengineering and Bioscience, Georgia Institute of Technology Atlanta, GA 30332
(USA)
| |
Collapse
|
62
|
Ahmed B, Sultana R, Greene MW. Adipose tissue and insulin resistance in obese. Biomed Pharmacother 2021; 137:111315. [PMID: 33561645 DOI: 10.1016/j.biopha.2021.111315] [Citation(s) in RCA: 290] [Impact Index Per Article: 96.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 02/08/2023] Open
Abstract
Currently, obesity has become a global health issue and is referred to as an epidemic. Dysfunctional obese adipose tissue plays a pivotal role in the development of insulin resistance. However, the mechanism of how dysfunctional obese-adipose tissue develops insulin-resistant circumstances remains poorly understood. Therefore, this review attempts to highlight the potential mechanisms behind obesity-associated insulin resistance. Multiple risk factors are directly or indirectly associated with the increased risk of obesity; among them, environmental factors, genetics, aging, gut microbiota, and diets are prominent. Once an individual becomes obese, adipocytes increase in their size; therefore, adipose tissues become larger and dysfunctional, recruit macrophages, and then these polarize to pro-inflammatory states. Enlarged adipose tissues release excess free fatty acids (FFAs), reactive oxygen species (ROS), and pro-inflammatory cytokines. Excess systemic FFAs and dietary lipids enter inside the cells of non-adipose organs such as the liver, muscle, and pancreas, and are deposited as ectopic fat, generating lipotoxicity. Toxic lipids dysregulate cellular organelles, e.g., mitochondria, endoplasmic reticulum, and lysosomes. Dysregulated organelles release excess ROS and pro-inflammation, resulting in systemic inflammation. Long term low-grade systemic inflammation prevents insulin from its action in the insulin signaling pathway, disrupts glucose homeostasis, and results in systemic dysregulation. Overall, long-term obesity and overnutrition develop into insulin resistance and chronic low-grade systemic inflammation through lipotoxicity, creating the circumstances to develop clinical conditions. This review also shows that the liver is the most sensitive organ undergoing insulin impairment faster than other organs, and thus, hepatic insulin resistance is the primary event that leads to the subsequent development of peripheral tissue insulin resistance.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Department of Nutrition, Auburn University, Auburn, AL, 36849, United States.
| | - Rifat Sultana
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, United States
| | - Michael W Greene
- Department of Nutrition, Auburn University, Auburn, AL, 36849, United States
| |
Collapse
|
63
|
Muñoz-Pérez VM, Ortiz MI, Salas-Casa A, Pérez-Guerrero J, Castillo-Pacheco N, Barragán-Ramírez G, Hernándes-Alejandro M. In vitro effects of citral on the human myometrium: Potential adjunct therapy to prevent preterm births. Birth Defects Res 2021; 113:613-622. [PMID: 33484091 DOI: 10.1002/bdr2.1873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Premature infants contribute to infant morbidity and mortality especially in low resource settings. Information on tocolytic and/or anti-inflammatory effects of several plant extracts, such as citral, could help prevent preterm birth cases and reduce the number of preterm infants. The aim of this study was to evaluate the in vitro tocolytic and anti-inflammatory effect of citral on myometrial tissues of the human uterus. METHODS Myometrial samples from uteri obtained after hysterectomy were used in functional tests to evaluate the inhibitory effect of citral on PGF-2α induced contractions. The intracellular cyclic adenosine monophosphate (cAMP) levels generated in response to citral in human myometrial homogenates were measured by ELISAs. Forskolin was used as a positive control. The anti-inflammatory effect of citral was determined through the measurement of two pro-inflammatory cytokines, tumor necrosis factor-α (TNFα) and interleukin (IL)-1β, and the anti-inflammatory cytokine IL-10, in human myometrial explants stimulated with lipopolysaccharide (LPS). RESULTS Citral was able to induce a significant inhibition of PGF-2α induced contractions at the highest concentration level (p < .05). Citral caused a concentration-dependent increase in myometrial cAMP levels (p < .05) and a concentration-dependent decrease in LPS-induced TNFα and IL-1β production, while IL-10 production increased significantly (p < .05). The anti-inflammatory and tocolytic effects induced by citral could be associated with an increase in cAMP levels in human myometrial samples. CONCLUSION These properties place citral as a potentially safe and effective adjuvant agent in preterm birth cases, an obstetric and gynecological problem that requires urgent attention.
Collapse
Affiliation(s)
- Víctor Manuel Muñoz-Pérez
- Área Académica de Medicina del Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, Mexico
| | - Mario I Ortiz
- Área Académica de Medicina del Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, Mexico
| | - Andrés Salas-Casa
- Área Académica de Gerontología del Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, Mexico
| | - Jessica Pérez-Guerrero
- Departamento de Ginecología y Obstetricia del Hospital General de los SSH, Pachuca, Mexico
| | - Narmi Castillo-Pacheco
- Departamento de Ginecología y Obstetricia del Hospital General de los SSH, Pachuca, Mexico
| | | | - Mario Hernándes-Alejandro
- Departamento de Bioingeniería, Unidad profesional Interdisciplinaria de biotecnología del Instituto Politécnico Nacional (UPIBI-IPN), México City, Mexico
| |
Collapse
|
64
|
O’Brien P, Han G, Ganpathy P, Pitre S, Zhang Y, Ryan J, Sim PY, Harding SV, Gray R, Preedy VR, Sanders TAB, Corpe CP. Chronic Effects of a High Sucrose Diet on Murine Gastrointestinal Nutrient Sensor Gene and Protein Expression Levels and Lipid Metabolism. Int J Mol Sci 2020; 22:E137. [PMID: 33375525 PMCID: PMC7794826 DOI: 10.3390/ijms22010137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/25/2022] Open
Abstract
The gastrointestinal tract (GIT) plays a key role in regulating nutrient metabolism and appetite responses. This study aimed to identify changes in the GIT that are important in the development of diet related obesity and diabetes. GIT samples were obtained from C57BL/6J male mice chronically fed a control diet or a high sucrose diet (HSD) and analysed for changes in gene, protein and metabolite levels. In HSD mice, GIT expression levels of fat oxidation genes were reduced, and increased de novo lipogenesis was evident in ileum. Gene expression levels of the putative sugar sensor, slc5a4a and slc5a4b, and fat sensor, cd36, were downregulated in the small intestines of HSD mice. In HSD mice, there was also evidence of bacterial overgrowth and a lipopolysaccharide activated inflammatory pathway involving inducible nitric oxide synthase (iNOS). In Caco-2 cells, sucrose significantly increased the expression levels of the nos2, iNOS and nitric oxide (NO) gas levels. In conclusion, sucrose fed induced obesity/diabetes is associated with changes in GI macronutrient sensing, appetite regulation and nutrient metabolism and intestinal microflora. These may be important drivers, and thus therapeutic targets, of diet-related metabolic disease.
Collapse
Affiliation(s)
- Patrick O’Brien
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - Ge Han
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - Priya Ganpathy
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - Shweta Pitre
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - Yi Zhang
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - John Ryan
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - Pei Ying Sim
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - Scott V. Harding
- Department of Biochemistry, Memorial University, Elizabeth Avenue, St. John’s, NL A1C5S7, Canada;
| | - Robert Gray
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - Victor R. Preedy
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - Thomas A. B. Sanders
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| | - Christopher P. Corpe
- Nutritional Sciences Division, Faculty of Life Sciences and Medicine, School of Life Courses, King’s College London, Room 3.114, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; (P.O.); (G.H.); (P.G.); (S.P.); (Y.Z.); (J.R.); (P.Y.S.); (R.G.); (V.R.P.); (T.A.B.S.)
| |
Collapse
|
65
|
Zhang C, Li CX, Shao Q, Chen WB, Ma L, Xu WH, Li YX, Huang SC, Ma YB. Effects of Glycyrrhiza polysaccharide in diet on growth performance, serum antioxidant capacity, and biochemistry of broilers. Poult Sci 2020; 100:100927. [PMID: 33518321 PMCID: PMC7936193 DOI: 10.1016/j.psj.2020.12.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
In the present study, we analyzed the effects of Glycyrrhiza polysaccharide (GCP) on growth performance, serum antioxidant capacity, and biochemistry of broilers. A total of 600, one-day-old AA broilers randomly divided into 5 treatment groups with 6 replicate pens of 20 birds per cage received dietary supplementation with GCP (0, 200, 500, 1,000, and 1,500 mg/kg) for 42 d. The supplementation of GCP linearly decreased (P < 0.05) feed conversion rate on day 22 to 42. Dietary supplementation with GCP reduced (P < 0.05) serum total cholesterol on day 21 and 42 and linearly improved (P < 0.05) albumin and high-density lipoprotein cholesterol. Dietary supplementation with 1,000 or 1,500 mg/kg GCP significantly increased (P < 0.05) serum total superoxide dismutase (T-SOD) activity on day 21 and 42 and reduced (P < 0.05) serum malondialdehyde content on 21 d. Dietary supplementation with 1,000 or 1,500 mg/kg GCP significantly improved (P < 0.05) interleukin-1β (IL-1β) and interferon-γ (IFN-γ) expressions in liver on day 21 and 42. At the end of the experiment, we randomly selected 20 broilers from 3 treatment groups (0, 1,000, and 1,500 mg/kg), respectively, to perform an lipopolysaccharide (LPS)-induced acute stress experiment. The 60 broilers were divided into 6 treatment groups with 10 birds per cage. The experiment was designed as a 3 × 2 factorial arrangement with GCP (0, 1,000, or 1,500 mg/kg) and LPS (injection of saline or 1 mg/kg body weight) levels as treatments. When the grouping was finished, the broilers were immediately intraperitoneally injected with LPS or normal saline. Six hours after challenged, serum antioxidant and liver immunity were analyzed. The results showed that dietary GCP prevented LPS-induced reductions in T-SOD activity and increases in malonaldehyde content (P < 0.05). Also, dietary GCP supplementation mitigated the LPS-induced increase in IL-1β and IFN-γ in the liver. Supplementation with 1,500 mg/kg GCP showed the most optimal effect in broilers. GCP has the potential to be used as feed additive in broilers.
Collapse
Affiliation(s)
- C Zhang
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang 471023, PR China.
| | - C X Li
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Q Shao
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang 471023, PR China
| | - W B Chen
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang 471023, PR China
| | - L Ma
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang 471023, PR China
| | - W H Xu
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Y X Li
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang 471023, PR China
| | - S C Huang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, PR China
| | - Y B Ma
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang 471023, PR China
| |
Collapse
|
66
|
Stančič B, Qvarfordt B, Berglund MM, Brenden N, Sydow Bäckman M, Fransson M, Nordling S, Magnusson PU. The blood endothelial cell chamber - An innovative system to study immune responses in drug development. Int Immunopharmacol 2020; 90:107237. [PMID: 33310662 DOI: 10.1016/j.intimp.2020.107237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/04/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
The risk for adverse immune-mediated reactions, associated with the administration of certain immunotherapeutic agents, should be mitigated early. Infusion reactions to monoclonal antibodies and other biopharmaceuticals, known as cytokine release syndrome, can arise from the release of cytokines via the drug target cell, as well as the recruitment of immune effector cells. While several in vitro cytokine release assays have been proposed up to date, many of them lack important blood components, required for this response to occur. The blood endothelial cell chamber model is an in vitro assay, composed of freshly drawn human whole blood and cultured human primary endothelial cells. Herein, its potential to study the compatibility of immunotherapeutics with the human immune system was studied by evaluating three commercially available monoclonal antibodies and bacterial endotoxin lipopolysaccharide. We demonstrate that the anti-CD28 antibody TGN1412 displayed an adaptive cytokine release profile and a distinct IL-2 response, accompanied with increased CD3+ cell recruitment. Alemtuzumab exhibited a clear cytokine response with a mixed adaptive/innate source (IFNγ, TNFα and IL-6). Its immunosuppressive nature is observed in depleted CD3+ cells. Cetuximab, associated with low infusion reactions, showed a very low or absent stimulatory effect on proinflammatory cytokines. In contrast, bacterial endotoxin demonstrated a clear innate cytokine response, defined by TNFα, IL-6 and IL-1β release, accompanied with a strong recruitment of CD14+CD16+ cells. Therefore, the blood endothelial cell chamber model is presented as a valuable in vitro tool to investigate therapeutic monoclonal antibodies with respect to cytokine release and vascular immune cell recruitment.
Collapse
Affiliation(s)
- Brina Stančič
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden; Department of Molecular Biology, Universidad Autónoma de Madrid, and Department of Molecular Neuropathology, Center of Molecular Biology Severo Ochoa (UAM-CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Bodil Qvarfordt
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden
| | | | - Nina Brenden
- Swedish Orphan Biovitrum AB, Tomtebodavägen 23A, 112 76 Solna, Sweden
| | | | - Moa Fransson
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden
| | - Sofia Nordling
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsväg 20, 751 85, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
67
|
Kwon S, Shin S, Do M, Oh BH, Song Y, Bui VD, Lee ES, Jo DG, Cho YW, Kim DH, Park JH. Engineering approaches for effective therapeutic applications based on extracellular vesicles. J Control Release 2020; 330:15-30. [PMID: 33278480 DOI: 10.1016/j.jconrel.2020.11.062] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022]
Abstract
The biological significance of extracellular vesicles (EVs) as intercellular communication mediators has been increasingly revealed in a wide range of normal physiological processes and disease pathogenesis. In particular, regenerative and immunomodulatory EVs hold potential as innate biotherapeutics, whereas pathological EVs are considered therapeutic targets for inhibiting their bioactivity. Given their ability to transport functional cargos originating from the source cells to target cells, EVs can also be used as a therapeutic means to deliver drug molecules. This review aims to provide an updated overview of the key engineering approaches for better exploiting EVs in disease intervention. The emphasis is lying on the preconditioning methods for therapeutic EVs, drug loading and targeting technologies for carrier EVs, and activity control strategies for pathological EVs.
Collapse
Affiliation(s)
- Seunglee Kwon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sol Shin
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Byeong Hoon Oh
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yeari Song
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Van Dat Bui
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Eun Sook Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Dong-Gyu Jo
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea; Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; ExoStemTech Inc., Ansan 15588, Republic of Korea
| | - Yong Woo Cho
- ExoStemTech Inc., Ansan 15588, Republic of Korea; Department of Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea; Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon 16419, Republic of Korea; ExoStemTech Inc., Ansan 15588, Republic of Korea.
| |
Collapse
|
68
|
Roberts JL, Liu G, Darby TM, Fernandes LM, Diaz-Hernandez ME, Jones RM, Drissi H. Bifidobacterium adolescentis supplementation attenuates fracture-induced systemic sequelae. Biomed Pharmacother 2020; 132:110831. [PMID: 33022534 PMCID: PMC9979243 DOI: 10.1016/j.biopha.2020.110831] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
The gut microbiota is an important contributor to both health and disease. While previous studies have reported on the beneficial influences of the gut microbiota and probiotic supplementation on bone health, their role in recovery from skeletal injury and resultant systemic sequelae remains unexplored. This study aimed to determine the extent to which probiotics could modulate bone repair by dampening fracture-induced systemic inflammation. Our findings demonstrate that femur fracture induced an increase in gut permeability lasting up to 7 days after trauma before returning to basal levels. Strikingly, dietary supplementation with Bifidobacterium adolescentis augmented the tightening of the intestinal barrier, dampened the systemic inflammatory response to fracture, accelerated fracture callus cartilage remodeling, and elicited enhanced protection of the intact skeleton following fracture. Together, these data outline a mechanism whereby dietary supplementation with beneficial bacteria can be therapeutically targeted to prevent the systemic pathologies induced by femur fracture.
Collapse
Affiliation(s)
- Joseph L. Roberts
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA,Nutrition and Health Sciences Program, Emory University, Atlanta, GA, USA
| | - Guanglu Liu
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
| | - Trevor M. Darby
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Lorenzo M. Fernandes
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Rheinallt M. Jones
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA; Nutrition and Health Sciences Program, Emory University, Atlanta, GA, USA.
| |
Collapse
|
69
|
Monoclonal Antibody to CD14, TLR4, or CD11b: Impact of Epitope and Isotype Specificity on ROS Generation by Human Granulocytes and Monocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5708692. [PMID: 33294123 PMCID: PMC7700042 DOI: 10.1155/2020/5708692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/24/2020] [Accepted: 11/10/2020] [Indexed: 02/01/2023]
Abstract
Lipopolysaccharides (LPSs or endotoxins) from Gram-negative bacteria represent pathogen-associated molecular patterns (PAMPs) that are recognized by CD14 and Toll-like receptor 4 (TLR4). Lipopolysaccharides prime polymorphonuclear leukocytes (PMNs) for substantial production of reactive oxygen species (ROS) during its response to secondary stimuli such as chemoattractants or pathogens. The excessive ROS production can damage surrounding host tissues, thereby amplifying the inflammatory reaction caused by pathogens. Today, specific antibodies against CD14, TLR4, and CD11b are being used as the essential tools to elucidate the role of these receptors in acute inflammation and some of these antibodies have advised as therapeutic agents for clinical use. Because each antibody has two antigen-binding arms [F(ab′)2] and one Fc arm, its effect on cellular response is much more complicated rather than simple blockage of target receptor. In fact, IgG antibody, once bound to target receptor, engages Fc receptors γ (FcγRs) and thereby is able to activate the adaptive immune system. The consequences of antibody-dependent binary heterotypic association of CD14, TLR4, or CD11b with FcγRs as well as homotypic one on ROS production are not well elucidated. Moreover, the consequences of antigenic recognition of CD14, TLR4, or CD11b by specific F(ab′)2 fragments are not always investigated. In this review, we will discuss known mechanisms underlying the therapeutic efficiency of CD14, TLR4, and CD11b/CD18 antibodies with a focus on LPS-dependent ROS or cytokine production by PMNs or monocytes. The impacts of F(ab′)2 as well as antibody IgG subclasses (isotypes) in therapeutic efficiency or agonistic potency of known antibodies against abovementioned receptors are presented. We also pay attention to how the efficiency of different IgG antibody subclasses is modulated during LPS-induced inflammation and by production of priming agents such as interferon γ (IFN-γ). Our review reinforces the molecular targets and therapeutic approaches to amelioration of harmful consequences of excessive activation of human pattern recognition receptors.
Collapse
|
70
|
Evani SJ, Karna SLR, Seshu J, Leung KP. Pirfenidone regulates LPS mediated activation of neutrophils. Sci Rep 2020; 10:19936. [PMID: 33203891 PMCID: PMC7672086 DOI: 10.1038/s41598-020-76271-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 09/24/2020] [Indexed: 12/16/2022] Open
Abstract
Excessive inflammation or its absence may result in impaired wound healing. Neutrophils are among the first innate immune cells to arrive at the injury site. They participate in infection control and debris removal to initiate healing. If not timely resolved, neutrophils can cause excessive tissue inflammation and damage. Drugs with anti-inflammatory and anti-fibrotic effects are of promise for improving healing by balancing the primary defensive functions and excessive tissue damage actions. Of interest, pirfenidone (Pf), an FDA approved anti-fibrotic drug to treat idiopathic pulmonary fibrosis, has been shown to ameliorate inflammation in several animal models including mouse deep partial-thickness burn wounds. However, there is a lack of mechanistic insights into Pf drug action on inflammatory cells such as neutrophils. Here, we examined the treatment effects of Pf on LPS-stimulated neutrophils as a model of non-sterile inflammation. Firstly, Pf reduced chemotaxis and production of pro-inflammatory ROS, cytokines, and chemokines by LPS-activated neutrophils. Secondly, Pf increased anti-inflammatory IL-1RA and reduced neutrophil degranulation, phagocytosis, and NETosis. Thirdly, Pf affected downstream signaling kinases which might directly or indirectly influence neutrophil responses to LPS. In conclusion, the results suggest that Pf lessens the inflammatory phenotypes of LPS-activated neutrophils.
Collapse
Affiliation(s)
- Shankar J Evani
- Division of Combat Wound Repair, U.S. Army Institute of Surgical Research, 3698 Chambers Pass, Building 3610, JBSA Fort Sam Houston, San Antonio, TX, 78234-7767, USA
| | - S L Rajasekhar Karna
- Division of Combat Wound Repair, U.S. Army Institute of Surgical Research, 3698 Chambers Pass, Building 3610, JBSA Fort Sam Houston, San Antonio, TX, 78234-7767, USA
| | - Janakiram Seshu
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Kai P Leung
- Division of Combat Wound Repair, U.S. Army Institute of Surgical Research, 3698 Chambers Pass, Building 3610, JBSA Fort Sam Houston, San Antonio, TX, 78234-7767, USA.
| |
Collapse
|
71
|
Oh Y, Jeong H, Lim S, Hong J. Controlled Nitric Oxide Release Using Poly(lactic-co-glycolic acid) Nanoparticles for Anti-Inflammatory Effects. Biomacromolecules 2020; 21:4972-4979. [DOI: 10.1021/acs.biomac.0c01176] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Yoogyeong Oh
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyejoong Jeong
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sungmin Lim
- Division of Cardiology, Department of Internal Medicine, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jinkee Hong
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
72
|
Gugliandolo E, Crupi R, Biondi V, Licata P, Cuzzocrea S, Passantino A. Protective Effect of Silibinin on Lipopolysaccharide-Induced Inflammatory Responses in Equine Peripheral Blood Mononuclear Cells, an In Vitro Study. Animals (Basel) 2020; 10:ani10112022. [PMID: 33153060 PMCID: PMC7692186 DOI: 10.3390/ani10112022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Natural compounds are often an important source of biologically active molecules, which can find important applications in the treatment or pharmacological prevention of several pathologies. Silibinin is a natural polyphenolic flavonoid that is extracted from plant milk thistle, Silybum marianum. Silibinin has been reported to have antioxidant and immunomodulatory and anti-inflammatory activities. In horses, in particular, inflammation secondary to bacterial infection or translocation is one of the most frequent causes of morbidity and mortality. The aim of this study was to test the effect of silibinin on lipopolysaccharide (LPS)-induced inflammatory response in equine peripheral blood mononuclear cells (PBMCs). Taken together, our results showed an interesting prospective in therapeutic use of silibinin in equine inflammatory disease. Furthermore, the results from this study support the evidence of use equine PBMCs as an in vitro model to study inflammatory and immune response and for drug screening into the target specie. Abstract Although inflammation is an important physiological response, it plays a prominent role in several diseases across the mammalian species. In horses, in particular, inflammation secondary to bacterial infection or translocation is one of the most frequent causes of morbidity and mortality. Research in new molecules with anti-inflammatory and immunomodulatory proprieties and safe use profile is constantly an active field; natural compounds are an important source of molecules with peculiar properties such as antioxidants, anti-inflammatory and immune modulating. Silibinin, a natural polyphenolic flavonoid, extracted from plant milk thistle, Silybum marianum, has been reported to have actions such as antioxidant immunomodulatory and anti-inflammatory. The aim of this study was to test the effect of silibinin on lipopolysaccharide (LPS)-induced inflammatory response in equine peripheral blood mononuclear cells (PBMCs). Our results showed the protective effect of silibinin 10 μM and 50 μM in equine PBMCs stimulated with LPS. Silibilinin was able to prevent the LPS induced increased levels of TNF-α, IL-1β, IL-6 and IL-8. The results from this study on LPS-stimulated equine PBMCs showed that silibinin could be a useful pharmacological approach in treatment or prevention of several inflammatory conditions in horse.
Collapse
Affiliation(s)
- Enrico Gugliandolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy;
| | - Rosalia Crupi
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy; (R.C.); (V.B.); (P.L.); (A.P.)
| | - Vito Biondi
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy; (R.C.); (V.B.); (P.L.); (A.P.)
| | - Patrizia Licata
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy; (R.C.); (V.B.); (P.L.); (A.P.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy;
- Department of Pharmacological and Physiological Science, Saint Louis University, School of Medicine, 1402 South Grand Blvd, St Louis, MO 63104, USA
- Correspondence:
| | - Annamaria Passantino
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy; (R.C.); (V.B.); (P.L.); (A.P.)
| |
Collapse
|
73
|
Crezee T, Petrulea M, Piciu D, Jaeger M, Smit JWA, Plantinga TS, Georgescu CE, Netea-Maier R. Akt1 genetic variants confer increased susceptibility to thyroid cancer. Endocr Connect 2020; 9:1065-1074. [PMID: 33112820 PMCID: PMC7774771 DOI: 10.1530/ec-20-0311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/02/2020] [Indexed: 11/30/2022]
Abstract
The PI3K-Akt-mTOR pathway plays a central role in the development of non-medullary thyroid carcinoma (NMTC). Although somatic mutations have been identified in these genes in NMTC patients, the role of germline variants has not been investigated. Here, we selected frequently occurring genetic variants in AKT1, AKT2, AKT3, PIK3CA and MTOR and have assessed their effect on NMTC susceptibility, progression and clinical outcome in a Dutch discovery cohort (154 patients, 188 controls) and a Romanian validation cohort (159 patients, 260 controls). Significant associations with NMTC susceptibility were observed for AKT1 polymorphisms rs3803304, rs2494732 and rs2498804 in the Dutch discovery cohort, of which the AKT1 rs3803304 association was confirmed in the Romanian validation cohort. No associations were observed between PI3K-Akt-mTOR polymorphisms and clinical parameters including histology, TNM staging, treatment response and clinical outcome. Functionally, cells bearing the associated AKT1 rs3803304 risk allele exhibit increased levels of phosphorylated Akt protein, potentially leading to elevated signaling activity of the oncogenic Akt pathway. All together, germline encoded polymorphisms in the PI3K-Akt-mTOR pathway could represent important risk factors in development of NMTC.
Collapse
Affiliation(s)
- Thomas Crezee
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mirela Petrulea
- Department of Endocrinology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Doina Piciu
- Department of Endocrinology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin Jaeger
- Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Nuclear Medicine and Endocrine Tumors, Institute of Oncology ‘Prof. Dr. Ion Chiricuta’, Cluj-Napoca, Romania
| | - Jan W A Smit
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Theo S Plantinga
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Nuclear Medicine and Endocrine Tumors, Institute of Oncology ‘Prof. Dr. Ion Chiricuta’, Cluj-Napoca, Romania
| | - Carmen E Georgescu
- Department of Endocrinology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Endocrinology Clinic, Cluj County Emergency Hospital, Cluj-Napoca, Romania
| | - Romana Netea-Maier
- Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Nuclear Medicine and Endocrine Tumors, Institute of Oncology ‘Prof. Dr. Ion Chiricuta’, Cluj-Napoca, Romania
- Correspondence should be addressed to R Netea-Maier:
| |
Collapse
|
74
|
Klann E, Tagliamonte MS, Ukhanova M, Mai V, Vedam-Mai V. Gut Microbiota Dynamics in Parkinsonian Mice. ACS Chem Neurosci 2020; 11:3267-3276. [PMID: 32941730 DOI: 10.1021/acschemneuro.0c00386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peripheral immunity is thought to be dysregulated in Parkinson's disease (PD) and may provide an avenue for novel immunotherapeutic interventions. Gut microbiota is a potential factor for modulating immunotherapy response. Considering the possibly complex role of the gut-brain axis in PD, we used a preclinical model to determine the effects of gut microbiota dynamics in mice receiving an immunotherapeutic intervention compared to controls. A total of 17 M83 heterozygous transgenic mice were used in this study. Mice in the treatment arm (N = 10) received adoptive cellular therapy (ACT) by injection, and control mice (N = 7) were injected with saline at 8 weeks of age. All mice received peripheral α-syn fibrils to hasten parkinsonian symptoms via an intramuscular injection 1 week later (9 weeks of age; baseline). Fecal pellets were collected from all mice at three time points postinjection (baseline, 6 weeks, and 12 weeks). DNA from each stool sample was extracted, and 16S rDNA was amplified, sequenced, and analyzed using QIIME2 and RStudio. Differences in the relative abundance of bacterial taxa were observed over time between groups. No significant differences in alpha diversity were found between groups at any time point. UniFrac measures of phylogenetic distance between samples demonstrated distinct clustering between groups postbaseline (p = 0.002). These differences suggest that the gut microbiome may be capable of influencing immunotherapy outcomes. Conclusively, we observed distinctly different microbiota dynamics in treated mice compared to those in the control group. These results suggest a correlation between the gut-brain axis, PD pathology, and immunotherapy.
Collapse
Affiliation(s)
- Emily Klann
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida 32611, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Massimiliano S. Tagliamonte
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida 32611, United States
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Maria Ukhanova
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Volker Mai
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Vinata Vedam-Mai
- Department of Neurology, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
75
|
Cromarty R, Sigal A, Liebenberg LJ, Mckinnon LR, Abdool Karim SS, Passmore JAS, Archary D. Betamethasone induces potent immunosuppression and reduces HIV infection in a PBMC in vitro model. J Investig Med 2020; 69:28-40. [PMID: 33004468 PMCID: PMC7803916 DOI: 10.1136/jim-2020-001424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 01/15/2023]
Abstract
Genital inflammation is an established risk factor for increased HIV acquisition risk. Certain HIV-exposed seronegative populations, who are naturally resistant to HIV infection, have an immune quiescent phenotype defined by reduced immune activation and inflammatory cytokines at the genital tract. Therefore, the aim of this study was to create an immune quiescent environment using immunomodulatory drugs to mitigate HIV infection. Using an in vitro peripheral blood mononuclear cell (PBMC) model, we found that inflammation was induced using phytohemagglutinin and Toll-like receptor (TLR) agonists Pam3CSK4 (TLR1/2), lipopolysaccharide (LPS) (TLR4) and R848 (TLR7/8). After treatment with anti-inflammatory drugs, ibuprofen (IBF) and betamethasone (BMS), PBMCs were exposed to HIV NL4-3 AD8. Multiplexed ELISA was used to measure 28 cytokines to assess inflammation. Flow cytometry was used to measure immune activation (CD38, HLA-DR and CCR5) and HIV infection (p24 production) of CD4+ T cells. BMS potently suppressed inflammation (soluble cytokines, p<0.05) and immune activation (CD4+ T cells, p<0.05). BMS significantly reduced HIV infection of CD4+ T cells only in the LPS (0.98%) and unstimulated (1.7%) conditions (p<0.02). In contrast, IBF had minimal anti-inflammatory and immunosuppressive but no anti-HIV effects. BMS demonstrated potent anti-inflammatory effects, regardless of stimulation condition. Despite uniform immunosuppression, BMS differentially affected HIV infection according to the stimulation conditions, highlighting the complex nature of these interactions. Together, these data underscore the importance of interrogating inflammatory signaling pathways to identify novel drug targets to mitigate HIV infection.
Collapse
Affiliation(s)
- Ross Cromarty
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
| | - Alexander Sigal
- Africa Health Research Institute (AHRI), Durban, KwaZulu-Natal, South Africa
- Max-Planck-Institute for Infection Biology, Berlin, Germany
| | - Lenine Julie Liebenberg
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Lyle Robert Mckinnon
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Salim Safurdeen Abdool Karim
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Jo-Ann Shelly Passmore
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Institute of Infectious Diseases and Molecular Medicine, University of Cape Town Faculty of Health Sciences, Cape Town, Western Cape, South Africa
| | - Derseree Archary
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
76
|
Bastin A, Sadeghi A, Nematollahi MH, Abolhassani M, Mohammadi A, Akbari H. The effects of malvidin on oxidative stress parameters and inflammatory cytokines in LPS-induced human THP-1 cells. J Cell Physiol 2020; 236:2790-2799. [PMID: 32914418 DOI: 10.1002/jcp.30049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 01/29/2023]
Abstract
Malvidin is an anthocyanin which is involved in inhibiting inflammatory-related mediators in inflammatory diseases; however, its mechanism of action in THP-1 cells is not yet known. THP-1 is a human monocytic cell line that is derived from patients with acute monocytic leukemia. The present study aimed to investigate the effect of malvidin on inflammatory responses and oxidative stress in lipopolysaccharide (LPS)-induced THP-1 cells. THP-1 cells were stimulated with LPS (50 ng/ml) to induce inflammation in the presence or absence of malvidin. The anti/proinflammatory cytokines were evaluated by real-time polymerase chain reaction and enzyme-linked immunosorbent assay. Total protein levels/phosphorylation of c-Jun N-terminal kinase (JNK), P65-NF-κB, and IKKα/IKKβ were evaluated by western blot analysis. Malondialdehyde (MDA) and nitric oxide (NO) metabolite levels, ferric reducing antioxidant power (FRAP), total thiol (T-SH) content, and superoxide dismutase (SOD) and glutathione peroxidase (GPx) activity were measured to evaluate the antioxidant activity of malvidin in THP-1 cells. Treatment of LPS-stimulated THP-1 cells with malvidin (100 and 200 μM) led to the significant inhibition of interleukin-6 (IL-6), tumor necrosis factor-α, and IL-1β messenger RNA (mRNA) expression and protein levels as well as a significant increase in the IL-10 mRNA expression and protein secretion. Moreover, 200 μM malvidin treatment reduced the phosphorylation of JNK, IKKα/IKKβ, and P65-NF-κB. These findings showed that malvidin not only decreased the MDA and NO metabolite levels but also increased the FRAP and T-SH content as well as SOD and GPx activities. The findings of the present study demonstrated the potential role of malvidin in blocking inflammation and oxidative stress induced by LPS in THP-1 cell line, suggesting that malvidin is likely to be a therapeutic agent for inflammatory diseases.
Collapse
Affiliation(s)
- Alireza Bastin
- Herbal and Traditional Medicine Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Asie Sadeghi
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hadi Nematollahi
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Moslem Abolhassani
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abbas Mohammadi
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamed Akbari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
77
|
Li ZM, Xu SY, Feng YZ, Cheng YR, Xiong JB, Zhou Y, Guan CX. The role of NOX4 in pulmonary diseases. J Cell Physiol 2020; 236:1628-1637. [PMID: 32780450 DOI: 10.1002/jcp.30005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/26/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
Nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) is a subtype of the NOX family, which is mainly expressed in the pulmonary vasculature and pulmonary endothelial cells in the respiratory system. NOX4 has unique characteristics, and is a constitutively active enzyme that primarily produces hydrogen peroxide. The signaling pathways associated with NOX4 are complicated. Negative and positive feedback play significant roles in regulating NOX4 expression. The role of NOX4 is controversial because NOX4 plays a protective or damaging role in different respiratory diseases. This review summarizes the structure, enzymatic properties, regulation, and signaling pathways of NOX4. This review then introduces the roles of NOX4 in different diseases in the respiratory system, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease, and pulmonary fibrosis.
Collapse
Affiliation(s)
- Zi-Ming Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Sheng-Ya Xu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yi-Zhuo Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yu-Rui Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
78
|
Hu J, Wang H, Li X, Liu Y, Mi Y, Kong H, Xi D, Yan W, Luo X, Ning Q, Wang X. Fibrinogen-like protein 2 aggravates nonalcoholic steatohepatitis via interaction with TLR4, eliciting inflammation in macrophages and inducing hepatic lipid metabolism disorder. Theranostics 2020; 10:9702-9720. [PMID: 32863955 PMCID: PMC7449923 DOI: 10.7150/thno.44297] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 07/24/2020] [Indexed: 12/17/2022] Open
Abstract
Rationale: The functions of fibrinogen-like protein 2 (fgl2) have been studied in many inflammatory and neoplastic diseases, but the role of fgl2 in nonalcoholic fatty liver disease has not yet been elucidated. In this study, we sought to investigate the role of fgl2 in the pathogenesis of nonalcoholic steatohepatitis (NASH). Methods: Hepatic fgl2 expression was tested in patients with nonalcoholic fatty liver (NAFL) or NASH and controls. Wild-type and fgl2-/- C57BL/6 mice were subjected to a methionine/choline-deficient (MCD) diet or a high-fat diet (HFD) to establish NASH models. Bone marrow-derived macrophages (BMDMs) stimulated with LPS or free fatty acids were used for the in vitro study. Results: In both humans and mice with NASH, macrophage accumulation was concomitant with significantly increased fgl2 expression in the liver. Fgl2 deficiency attenuated liver steatosis and inflammation in diet-induced murine models of NASH. In both liver tissues and BMDMs from NASH mice, fgl2 deficiency resulted in reduced levels of proinflammatory cytokines and reactive oxygen species (ROS) compared with levels in wild-type controls. Activation of NF-κB, p38-MAPK and NLRP3 inflammasomes was also suppressed upon fgl2 disruption. Moreover, lipogenic genes (Fasn and SREBP-2) were downregulated while lipolytic genes (PPAR and CPT1A) were upregulated in the livers of fgl2-/- NASH mice. Primary hepatocytes incubated with the medium collected from fgl2-/- BMDMs showed less fat deposition than those incubated with WT BMDMs. Furthermore, we discovered that fgl2 combined with TLR4 mediates the activation of the Myd88-dependent signaling pathway, which may contribute to inflammation and lipid metabolism disorders. Conclusions: These data suggest that fgl2 aggravates the progression of NASH through activation of NF-κB, p38-MAPK and NLRP3 inflammasomes in macrophages, which consequently induces overproduction of proinflammatory cytokines and lipid metabolism disorders. An interaction of fgl2 and TLR4 may in part contribute to the activation of inflammatory signaling pathways in macrophages.
Collapse
Affiliation(s)
- Junjian Hu
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongwu Wang
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xitang Li
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yonggang Liu
- Tianjin Second People's Hospital and Tianjin Institute of Hepatology, Tianjin, China
| | - Yuqiang Mi
- Tianjin Second People's Hospital and Tianjin Institute of Hepatology, Tianjin, China
| | - Hongyan Kong
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dong Xi
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weiming Yan
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoping Luo
- Department and institute of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Ning
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaojing Wang
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
79
|
Nam U, Kim S, Park J, Jeon JS. Lipopolysaccharide-Induced Vascular Inflammation Model on Microfluidic Chip. MICROMACHINES 2020; 11:mi11080747. [PMID: 32751936 PMCID: PMC7465530 DOI: 10.3390/mi11080747] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
Inflammation is the initiation of defense of our body against harmful stimuli. Lipopolysaccharide (LPS), originating from outer membrane of Gram-negative bacteria, causes inflammation in the animal’s body and can develop several diseases. In order to study the inflammatory response to LPS of blood vessels in vitro, 2D models have been mainly used previously. In this study, a microfluidic device was used to investigate independent inflammatory response of endothelial cells by LPS and interaction of inflamed blood vessel with monocytic THP-1 cells. Firstly, the diffusion of LPS across the collagen gel into blood vessel was simulated using COMSOL. Then, inflammatory response to LPS in engineered blood vessel was confirmed by the expression of Intercellular Adhesion Molecule 1 (ICAM-1) and VE-cadherin of blood vessel, and THP-1 cell adhesion and migration assay. Upregulation of ICAM-1 and downregulation of VE-cadherin in an LPS-treated condition was observed compared to normal condition. In the THP-1 cell adhesion and migration assay, the number of adhered and trans-endothelial migrated THP-1 cells were not different between conditions. However, migration distance of THP-1 was longer in the LPS treatment condition. In conclusion, we recapitulated the inflammatory response of blood vessels and the interaction of THP-1 cells with blood vessels due to the diffusion of LPS.
Collapse
Affiliation(s)
- Ungsig Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141S, Korea; (U.N.); (S.K.); (J.P.)
| | - Seunggyu Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141S, Korea; (U.N.); (S.K.); (J.P.)
| | - Joonha Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141S, Korea; (U.N.); (S.K.); (J.P.)
| | - Jessie S. Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141S, Korea; (U.N.); (S.K.); (J.P.)
- KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
- Correspondence: ; Tel.: +82-42-350-3226
| |
Collapse
|
80
|
Modulatory Effect of Nicotinic Acid on the Metabolism of Caco-2 Cells Exposed to IL-1β and LPS. Metabolites 2020; 10:metabo10050204. [PMID: 32429415 PMCID: PMC7281454 DOI: 10.3390/metabo10050204] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/27/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are the most common gastrointestinal inflammatory pathologies. Previous work evidenced a lower content of nicotinic acid (NA) in feces of IBD patients compared to healthy subjects. In the present study, we aimed to understand the effects of NA on intestinal inflammation, as several studies reported its possible beneficial effect, and investigate its influence on inflammation-driven metabolism. NA was tested on a Caco-2 in-vitro model in which inflammation was induced with interleukin-1β (IL-1β) and lipopolysaccharide (LPS), two mayor proinflammatory compounds produced in IBD, that stimulate the production of cytokines, such as interleukin 8. A metabolomics approach, with gas chromatography–mass spectrometry (GC-MS) and nuclear proton magnetic resonance (1H-NMR), was applied to study the metabolic changes. The results showed that NA significantly reduced the level of IL-8 produced in both LPS and IL-1β stimulated cells, confirming the anti-inflammatory effect of NA also on intestinal inflammation. Moreover, it was demonstrated that NA treatment had a restoring effect on several metabolites whose levels were modified by treatments with IL-1β or LPS. This study points out a possible use of NA as anti-inflammatory compound and might be considered as a promising starting point in understanding the beneficial effect of NA in IBD.
Collapse
|
81
|
Stephens M, von der Weid PY. Lipopolysaccharides modulate intestinal epithelial permeability and inflammation in a species-specific manner. Gut Microbes 2020; 11:421-432. [PMID: 31203717 PMCID: PMC7524286 DOI: 10.1080/19490976.2019.1629235] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Patients presenting with Inflammatory bowel disease have been shown to exhibit an altered microbiome in both Crohn's disease and Ulcerative colitis. This shift in the microbial content led us to question whether several of these microbes are important in inflammatory processes present in these diseases and more specifically whether lipopolysaccharides from the gram-negative cell wall differentially stimulates resident cells. We, therefore, investigated the possible contribution of five major species of gram-negative bacteria found to be altered in presence during disease progression and evaluate their pathogenicity through LPS. We demonstrated that LPS from these different species had individual capacities to induce NF-κB and pro-inflammatory IL-8 production from HEK-TLR4 cells in a TLR4 dependent manner. Additional work using human intestinal colonic epithelial cell monolayers (Caco-2) demonstrated that the cells responded to the serotype specific LPS in a distinct manner, inducing many inflammatory mediators such as TNF-α and IL-10 in significantly altered proportions. Furthermore, the permeability of Caco-2 monolayers, as a test for their ability to alter intestinal permeability, was also differentially altered by the serotype specific LPS modulating trans-epithelial electrical resistance, small molecule movement, and tight junction integrity. Our results suggest that specific species of bacteria may be potentiating the pathogenesis of IBD and chronic inflammatory diseases through their serotype specific LPS responses.
Collapse
Affiliation(s)
- Matthew Stephens
- Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Pierre-Yves von der Weid
- Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada,CONTACT Pierre-Yves von der Weid Snyder Institute of Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
82
|
Tang FK, Yu ZH, Wong THF, Chung CYS, Hirao H, Au-Yeung HY. Fluorescein-Containing Superoxide Probes with a Modular Copper-Based Trigger. Chempluschem 2020; 85:653-658. [PMID: 32237224 DOI: 10.1002/cplu.202000059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/13/2020] [Indexed: 12/22/2022]
Abstract
Fluorescein-derived superoxide probes featuring a copper(II) complex that can be activated by superoxide to initiate ether bond cleavage and uncage a fluorescein reporter for imaging in live cells are described. Compared to other superoxide sensing moieties, this bond cleavage strategy can be modularly adapted to fluorescent reporters with different properties without compromising the superoxide reactivity and selectivity. A green-emitting probe and its lysosome-targeting analogue have been successfully developed. Both probes are sensitive with more than 30-fold fluorescence enhancement towards superoxide and are highly selective with no significant response towards other reactive oxygen species. A structure-activity relationship study of the copper-based superoxide trigger showed that the secondary coordination environment of the copper(II) center is important for the superoxide reactivity and selectivity. The probes have been applied in imaging changes in intracellular superoxide level in live HeLa and HEK293T cells upon menadione stimulation and also in a cellular inflammation model in RAW 264.7 cells.
Collapse
Affiliation(s)
- Fung Kit Tang
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| | - Zuo Hang Yu
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| | - Thomas Hin-Fung Wong
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| | - Clive Yik-Sham Chung
- Department of Chemistry, University of California, Berkeley, California, CA, 94720, USA
| | - Hajime Hirao
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| | - Ho Yu Au-Yeung
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| |
Collapse
|
83
|
Antidepressant-like effects of β-caryophyllene on restraint plus stress-induced depression. Behav Brain Res 2019; 380:112439. [PMID: 31862467 DOI: 10.1016/j.bbr.2019.112439] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/15/2019] [Accepted: 12/15/2019] [Indexed: 01/02/2023]
Abstract
Chronic stress is depressogenic by altering neurotrophic and neuroinflammatory environments of the organism. The endocannabinoid system controls cognitive and emotional responses related with stress through the interaction with endocannabinoid receptors. β-Caryophyllene (BCP) is a CB2 agonist that exhibited anti-inflammatory, analgesic effects but minimal psychoactive effects. To test if BCP exhibits antidepressant-like action, animals were chronically restrained with additional stressors for 28 days, and BCP (25, 50, 100 mg/kg) was intraperitoneally injected once a day during the stress inflicting period. Then despair related behaviors and hippocampal expression of neurotrophic, inflammatory and cannabinoid receptor levels were measured. To test the effect of BCP on long-term depression, field potentials were measured during the application of lipopolysaccharide and low frequency stimulation. In the tail suspension test and forced swim test, chronic stress-induced despair behaviors were reduced by BCP. Also BCP improved the stress-related changes in the hippocampal expression of COX-2, BDNF, and CB2 receptor expression. In organotypic hippocampal slices, BCP reduced the lipopolysaccharide-induced intensification of the long-term depression. In conclusion, BCP improved chronic stress related behavioral and biochemical changes. These results suggest that BCP may be effective in treating depression and stress related mental illnesses.
Collapse
|
84
|
Isolation and identification of anti-periodontitis ingredients in Lactobacillus paracasei subsp. paracasei NTU 101-fermented skim milk in vitro. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
85
|
Elmarakby AA, Ibrahim AS, Katary MA, Elsherbiny NM, El-Shafey M, Abd-Elrazik AM, Abdelsayed RA, Maddipati KR, Al-Shabrawey M. A dual role of 12/15-lipoxygenase in LPS-induced acute renal inflammation and injury. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1669-1680. [PMID: 31349026 DOI: 10.1016/j.bbalip.2019.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/17/2019] [Accepted: 07/19/2019] [Indexed: 01/06/2023]
Abstract
Recent studies suggest a potential role of bioactive lipids in acute kidney injury induced by lipopolysaccharide (LPS). The current study was designed to determine the profiling activities of various polyunsaturated fatty acid (PUFA) metabolizing enzymes, including lipoxygenases (LO), cyclooxygenase, and cytochrome P450 in the plasma of LPS-injected mice using LC-MS. Heat map analysis revealed that out of 126 bioactive lipids screened, only the 12/15-LO metabolite, 12-HETE, had a significant (2.24 ± 0.4) fold increase relative to control (P = 0.0001) after Bonferroni Correction (BCF α = 0.003). We then determined the role of the 12/15-LO in LPS-induced acute kidney injury using genetic and pharmacological approaches. Treatment of LPS injected mice with the 12/15-LO inhibitor, baicalein, significantly reduced levels of renal injury and inflammation markers including urinary thiobarbituric acid reactive substance (TBARs), urinary monocyte chemoattractant protein-1 (MCP-1), renal interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). Similarly, knocking-out of 12/15-LO reduced levels of renal inflammation and injury markers elicited by LPS injection. Next, we tested whether exogenous supplementation with docosahexaenoic acid (DHA) as a substrate would divert the role of 12/15-LO from being pro-inflammatory to anti-inflammatory via increased production of the anti-inflammatory metabolite. DHA treatment restored the decreased in plasma level of resolvin D2 (RvD2) and reduced renal injury in LPS-injected mice whereas DHA treatment failed to provide any synergistic effects in reducing renal injury in LPS injected 12/15-LO knock-out mice. The ability of RvD2 to protect kidney against LPS-induced renal injury was further confirmed by exogenous RvD2 which significantly reduced the elevation in renal injury in LPS injected mice. These data suggest a double-edged sword role of 12/15-LO in LPS-induced acute renal inflammation and injury, depending on the type of substrate available for its activity.
Collapse
Affiliation(s)
- Ahmed A Elmarakby
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA; Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Egypt.
| | - Ahmed S Ibrahim
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt; Wayne State University, Department of Ophthalmology, Visual, and Anatomical Sciences, Department of Pharmacology, Detroit, MI
| | - Mohamed A Katary
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA; Department of Pharmacology, Faculty of Pharmacy, Damnhour University, Egypt
| | - Nehal M Elsherbiny
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Shafey
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA; Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed M Abd-Elrazik
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA
| | - Rafik A Abdelsayed
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA
| | | | - Mohamed Al-Shabrawey
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA, USA; Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt; Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA.
| |
Collapse
|
86
|
Cromarty R, Sigal A, Liebenberg LJP, McKinnon LR, Abdool Karim SS, Passmore JAS, Archary D. Diminished HIV Infection of Target CD4+ T Cells in a Toll-Like Receptor 4 Stimulated in vitro Model. Front Immunol 2019; 10:1705. [PMID: 31396221 PMCID: PMC6664077 DOI: 10.3389/fimmu.2019.01705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
Genital inflammation is associated with increased HIV acquisition risk. Induction of an inflammatory response can occur through the recognition of pathogenic or commensal microbes by Toll-like receptors (TLRs) on various immune cells. We used a in vitro peripheral blood mononuclear cell (PBMC) system to understand the contribution of TLR stimulation in inducing inflammation and the activation of target T cells, and its effect on HIV susceptibility. PBMCs were stimulated with TLR agonists LPS (TLR4), R848 (TLR7/8), and Pam3CSK4 (TLR1/2), and then infected with HIV NL4-3 AD8. Multiplexed ELISA was used to measure 28 cytokines in cell culture supernatants. Flow cytometry was used to measure the activation state (CD38 and HLA-DR), and CCR5 expression on CD4+ and CD8+ T cells. Although TLR agonists induced higher cytokine and chemokine secretion, they did not significantly activate CD4+ and CD8+ T cells and showed decreased CCR5 expression relative to the unstimulated control. Despite several classes of inflammatory cytokines and chemokines being upregulated by TLR agonists, CD4+ T cells were significantly less infectable by HIV after TLR4-stimulation than the unstimulated control. These data demonstrate that the inflammatory effects that occur in the presence TLR agonist stimulations do not necessarily translate to the activation of T cells. Most importantly, the finding that TLR4-stimulation reduces rather than increases susceptibility of CD4+ T cells to HIV infection in this in vitro system strongly suggests that the increased chemokine and possible antiviral factor expression induced by these TLR agonists play a powerful although complex role in determining HIV infection risk.
Collapse
Affiliation(s)
- Ross Cromarty
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Alex Sigal
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Lenine J P Liebenberg
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Lyle R McKinnon
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Jo-Ann S Passmore
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Medical School, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
87
|
Muñoz VL, Porsch EA, St Geme JW. Kingella kingae Surface Polysaccharides Promote Resistance to Neutrophil Phagocytosis and Killing. mBio 2019; 10:e00631-19. [PMID: 31239373 PMCID: PMC6593399 DOI: 10.1128/mbio.00631-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 05/22/2019] [Indexed: 11/30/2022] Open
Abstract
Bacterial pathogens have evolved strategies that enable them to evade neutrophil-mediated killing. The Gram-negative coccobacillus Kingella kingae is an emerging pediatric pathogen and is increasingly recognized as a common etiological agent of osteoarticular infections and bacteremia in young children. K. kingae produces a polysaccharide capsule and an exopolysaccharide, both of which are important for protection against complement-mediated lysis and are required for full virulence in an infant rat model of infection. In this study, we examined the role of the K. kingae polysaccharide capsule and exopolysaccharide in protection against neutrophil killing. In experiments with primary human neutrophils, we found that the capsule interfered with the neutrophil oxidative burst response and prevented neutrophil binding of K. kingae but had no effect on neutrophil internalization of K. kingae In contrast, the exopolysaccharide resisted the bactericidal effects of antimicrobial peptides and efficiently blocked neutrophil phagocytosis of K. kingae This work demonstrates that the K. kingae polysaccharide capsule and exopolysaccharide promote evasion of neutrophil-mediated killing through distinct yet complementary mechanisms, providing additional support for the K. kingae surface polysaccharides as potential vaccine antigens. In addition, these studies highlight a novel interplay between a bacterial capsule and a bacterial exopolysaccharide and reveal new properties for a bacterial exopolysaccharide, with potential applicability to other bacterial pathogens.IMPORTANCEKingella kingae is a Gram-negative commensal in the oropharynx and represents a leading cause of joint and bone infections in young children. The mechanisms by which K. kingae evades host innate immunity during pathogenesis of disease remain poorly understood. In this study, we established that the K. kingae polysaccharide capsule and exopolysaccharide function independently to protect K. kingae against reactive oxygen species (ROS) production, neutrophil phagocytosis, and antimicrobial peptides. These results demonstrate the intricacies of K. kingae innate immune evasion and provide valuable information that may facilitate development of a polysaccharide-based vaccine against K. kingae.
Collapse
Affiliation(s)
- Vanessa L Muñoz
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Eric A Porsch
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Joseph W St Geme
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
88
|
Akgün E, Lunzer MM, Portoghese PS. Combined Glia Inhibition and Opioid Receptor Agonism Afford Highly Potent Analgesics without Tolerance. ACS Chem Neurosci 2019; 10:2004-2011. [PMID: 30110531 DOI: 10.1021/acschemneuro.8b00323] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Commonly prescribed opioid analgesics produce tolerance upon chronic use due in part to induction of hyperalgesia. Given that two reported bivalent ligands (MMG22 and MCC22) produce potent antinociception without tolerance only in inflamed mice, we have investigated the possible cellular and receptor targets of these ligands. The selective microglia inhibitors, minocycline and SB290157, antagonized intrathecal (i.t.) MCC22 antinociception orders of magnitude more potently than MMG22, suggesting that MCC22 selectively targets activated microglia. The astrocyte toxin, l-α-aminoadipic acid antagonized MMG22 antinociception 126-fold without reducing the potency of MCC22, indicating that activated astrocytes are targets of MMG22. MK-801 and Ro25-6981 antagonism of MMG22 antinociception, but not MCC22, is consistent with selective inhibition of activated NMDAR in astrocytes. The antinociception produced by i.t. MMG22 or MCC22 were both antagonized by the selective mu opioid receptor antagonist, β-FNA, implicating interaction of these ligands with MOR in spinal afferent neurons. MCC22 antinociception was potently blocked by kainate or AMPA ion channel antagonists (LY382884; NBQX), in contrast to MMG22. It is concluded that i.t. MMG22 and MCC22 produce exceptional antinociception via potent inhibition of activated spinal glia, thereby leading to desensitization of spinal neurons and enhanced activation of neuronal MOR. Thus, the present study suggests a new approach to treatment of chronic inflammatory pain without tolerance through a single molecular entity that simultaneously inhibits activated glia and stimulates MOR in spinal neurons.
Collapse
Affiliation(s)
- Eyup Akgün
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary M. Lunzer
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Philip S. Portoghese
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
89
|
Magalhães CB, Casquilho NV, Machado MN, Riva DR, Travassos LH, Leal-Cardoso JH, Fortunato RS, Faffe DS, Zin WA. The anti-inflammatory and anti-oxidative actions of eugenol improve lipopolysaccharide-induced lung injury. Respir Physiol Neurobiol 2019; 259:30-36. [DOI: 10.1016/j.resp.2018.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/28/2018] [Accepted: 07/07/2018] [Indexed: 11/28/2022]
|
90
|
Rakshit A, Khatua K, Shanbhag V, Comba P, Datta A. Cu 2+ selective chelators relieve copper-induced oxidative stress in vivo. Chem Sci 2018; 9:7916-7930. [PMID: 30450181 PMCID: PMC6202919 DOI: 10.1039/c8sc04041a] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/29/2018] [Indexed: 02/06/2023] Open
Abstract
Copper ions are essential for biological function yet are severely detrimental when present in excess. At the molecular level, copper ions catalyze the production of hydroxyl radicals that can irreversibly alter essential bio-molecules. Hence, selective copper chelators that can remove excess copper ions and alleviate oxidative stress will help assuage copper-overload diseases. However, most currently available chelators are non-specific leading to multiple undesirable side-effects. The challenge is to build chelators that can bind to copper ions with high affinity but leave the levels of essential metal ions unaltered. Here we report the design and development of redox-state selective Cu ion chelators that have 108 times higher conditional stability constants toward Cu2+ compared to both Cu+ and other biologically relevant metal ions. This unique selectivity allows the specific removal of Cu2+ ions that would be available only under pathophysiological metal overload and oxidative stress conditions and provides access to effective removal of the aberrant redox-cycling Cu ion pool without affecting the essential non-redox cycling Cu+ labile pool. We have shown that the chelators provide distinct protection against copper-induced oxidative stress in vitro and in live cells via selective Cu2+ ion chelation. Notably, the chelators afford significant reduction in Cu-induced oxidative damage in Atp7a-/- Menkes disease model cells that have endogenously high levels of Cu ions. Finally, in vivo testing of our chelators in a live zebrafish larval model demonstrate their protective properties against copper-induced oxidative stress.
Collapse
Affiliation(s)
- Ananya Rakshit
- Department of Chemical Sciences , Tata Institute of Fundamental Research , 1 Homi Bhabha Road, Colaba , Mumbai-400005 , India .
| | - Kaustav Khatua
- Department of Chemical Sciences , Tata Institute of Fundamental Research , 1 Homi Bhabha Road, Colaba , Mumbai-400005 , India .
| | - Vinit Shanbhag
- Department of Biochemistry , Christopher S. Bond Life Science Center , University of Missouri , Columbia , USA
| | - Peter Comba
- Universität Heidelberg , Anorganisch-Chemisches Institut , Interdisciplinary Center for Scientific Computing , INF 270 , D-69120 Heidelberg , Germany
| | - Ankona Datta
- Department of Chemical Sciences , Tata Institute of Fundamental Research , 1 Homi Bhabha Road, Colaba , Mumbai-400005 , India .
| |
Collapse
|
91
|
Arimilli S, Schmidt E, Damratoski BE, Prasad GL. Role of Oxidative Stress in the Suppression of Immune Responses in Peripheral Blood Mononuclear Cells Exposed to Combustible Tobacco Product Preparation. Inflammation 2018; 40:1622-1630. [PMID: 28577134 PMCID: PMC5587635 DOI: 10.1007/s10753-017-0602-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cigarette smoking is a major risk factor for several human diseases. Chronic inflammation, resulting from increased oxidative stress, has been suggested as a mechanism that contributes to the increased susceptibility of smokers to cancer and microbial infections. We have previously shown that whole-smoke conditioned medium (WS-CM) and total particulate matter (TPM) prepared from Kentucky 3R4F reference cigarettes [collectively called as combustible tobacco product preparations (TPPs)] potently suppressed agonist-stimulated cytokine secretion and target cell killing in peripheral blood mononuclear cells (PBMCs). Here we have investigated the role of oxidative stress from TPPs, which alters inflammatory responses in vitro. Particularly, we investigated the mechanisms of WS-CM-induced suppression of select cytokine secretions in Toll-like receptor (TLR) agonist-stimulated cells and target cell killing by effector cells in PBMCs. Pretreatment with N-acetyl cysteine (NAC), a precursor of reduced glutathione and an established anti-oxidant, protected against DNA damage and cytotoxicity caused by exposure to WS-CM. Similarly, secretion of tumor necrosis factor (TNF), interleukin (IL)-6, and IL-8 in response to TLR-4 stimulation was restored by pretreatment with NAC. Target cell killing, a functional measure of cytolytic cells in PBMCs, is suppressed by WS-CM. Pretreatment with NAC restored the target cell killing in WS-CM treated PBMCs. This was accompanied by higher perforin levels in the effector cell populations. Collectively, these data suggest that reducing oxidative stress caused by cigarette smoke components restores select immune responses in this ex vivo model.
Collapse
Affiliation(s)
- Subhashini Arimilli
- Department of Microbiology & Immunology, Wake Forest University Health Sciences, Room 2N-052, 575 Patterson Avenue, Winston-Salem, NC, 27101, USA.
| | | | - Brad E Damratoski
- Department of Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - G L Prasad
- RAI Services Company, Winston-Salem, NC, USA
| |
Collapse
|
92
|
Zhang X, Mardinoglu A, Joosten LAB, Kuivenhoven JA, Li Y, Netea MG, Groen AK. Identification of Discriminating Metabolic Pathways and Metabolites in Human PBMCs Stimulated by Various Pathogenic Agents. Front Physiol 2018. [PMID: 29535640 PMCID: PMC5835230 DOI: 10.3389/fphys.2018.00139] [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] [Indexed: 11/13/2022] Open
Abstract
Immunity and cellular metabolism are tightly interconnected but it is not clear whether different pathogens elicit specific metabolic responses. To address this issue, we studied differential metabolic regulation in peripheral blood mononuclear cells (PBMCs) of healthy volunteers challenged by Candida albicans, Borrelia burgdorferi, lipopolysaccharide, and Mycobacterium tuberculosis in vitro. By integrating gene expression data of stimulated PBMCs of healthy individuals with the KEGG pathways, we identified both common and pathogen-specific regulated pathways depending on the time of incubation. At 4 h of incubation, pathogenic agents inhibited expression of genes involved in both the glycolysis and oxidative phosphorylation pathways. In contrast, at 24 h of incubation, particularly glycolysis was enhanced while genes involved in oxidative phosphorylation remained unaltered in the PBMCs. In general, differential gene expression was less pronounced at 4 h compared to 24 h of incubation. KEGG pathway analysis allowed differentiation between effects induced by Candida and bacterial stimuli. Application of genome-scale metabolic model further generated a Candida-specific set of 103 reporter metabolites (e.g., desmosterol) that might serve as biomarkers discriminating Candida-stimulated PBMCs from bacteria-stimuated PBMCs. Our analysis also identified a set of 49 metabolites that allowed discrimination between the effects of Borrelia burgdorferi, lipopolysaccharide and Mycobacterium tuberculosis. We conclude that analysis of pathogen-induced effects on PBMCs by a combination of KEGG pathways and genome-scale metabolic model provides deep insight in the metabolic changes coupled to host defense.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Adil Mardinoglu
- Department of Chemical and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.,Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands
| | - Jan A Kuivenhoven
- Section Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Yang Li
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Albert K Groen
- Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
93
|
Casquilho NV, Moreira-Gomes MD, Magalhães CB, Okuro RT, Ortenzi VH, Feitosa-Lima EK, Lima LM, Barreiro EJ, Soares RM, Azevedo SMFO, Valença SS, Fortunato RS, Carvalho AR, Zin WA. Oxidative imbalance in mice intoxicated by microcystin-LR can be minimized. Toxicon 2018; 144:75-82. [PMID: 29454806 DOI: 10.1016/j.toxicon.2018.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 12/19/2022]
Abstract
Microcystins-LR (MC-LR) is a cyanotoxin produced by cyanobacteria. We evaluated the antioxidant potential of LASSBio-596 (LB-596, inhibitor of phosphodiesterases 4 and 5), per os, and biochemical markers involved in lung and liver injury induced by exposure to sublethal dose of MC-LR. Fifty male Swiss mice received an intraperitoneal injection of 60 μL of saline (CTRL group, n = 20) or a sublethal dose of MC-LR (40 μg/kg, TOX group, n = 20). After 6 h the animals received either saline (TOX and CTRL groups) or LB-596 (50 mg/kg, TOX + LASS group, n = 10) by gavage. At 6 h after exposure, respiratory mechanics was evaluated in 10 CTRL and 10 TOX mice: there was a significant increase of all lung mechanics parameters (static elastance, viscoelastic component of elastance and lung resistive and viscoelastic/inhomogeneous pressures) in TOX compared to CTRL. 8 h after saline or MC-LR administration, i.e., 2 h after treatment with LB-596, blood serum levels of alanine aminotransferase and aspartate aminotransferase, activity of superoxide dismutase, catalase, and content of malondialdehyde and carbonyl in lung and liver, NADPH oxidase 2 and 4 mRNA expressions, dual oxidase enzyme activity and H2O2 generation were analyzed in lung homogenates. All parameters were significantly higher in TOX than in the other groups. There was no significant difference between CTRL and TOX + LASS. MC-LR deteriorated lung and liver functions and induced redox imbalance in them, which was prevented by oral administration of LB-596.
Collapse
Affiliation(s)
- Natália V Casquilho
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Diana Moreira-Gomes
- Laboratory of Respiratory Physiology and Biochemistry, Superior Institute of Biomedical Sciences, Universidade Estadual do Ceará, Fortaleza, Brazil
| | - Clarissa B Magalhães
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata T Okuro
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor Hugo Ortenzi
- Laboratory of Molecular Radiobiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emanuel K Feitosa-Lima
- Laboratory of Biology Redox, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lidia M Lima
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio(®)), Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eliezer J Barreiro
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio(®)), Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel M Soares
- Laboratory of Ecophysiology and Toxicology of Cyanobacteria, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; NUMPEX-BIO - Multidisciplinar Center of Biological Research, Universidade Federal do Rio de Janeiro, Polo Xerém, Duque de Caxias, RJ, Brazil
| | - Sandra M F O Azevedo
- Laboratory of Ecophysiology and Toxicology of Cyanobacteria, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Samuel S Valença
- Laboratory of Biology Redox, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo S Fortunato
- Laboratory of Molecular Radiobiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alysson Roncally Carvalho
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Walter A Zin
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
94
|
Pro-inflammatory hepatic macrophages generate ROS through NADPH oxidase 2 via endocytosis of monomeric TLR4-MD2 complex. Nat Commun 2017; 8:2247. [PMID: 29269727 PMCID: PMC5740170 DOI: 10.1038/s41467-017-02325-2] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) contribute to the development of non-alcoholic fatty liver disease. ROS generation by infiltrating macrophages involves multiple mechanisms, including Toll-like receptor 4 (TLR4)-mediated NADPH oxidase (NOX) activation. Here, we show that palmitate-stimulated CD11b+F4/80low hepatic infiltrating macrophages, but not CD11b+F4/80high Kupffer cells, generate ROS via dynamin-mediated endocytosis of TLR4 and NOX2, independently from MyD88 and TRIF. We demonstrate that differently from LPS-mediated dimerization of the TLR4–MD2 complex, palmitate binds a monomeric TLR4–MD2 complex that triggers endocytosis, ROS generation and increases pro-interleukin-1β expression in macrophages. Palmitate-induced ROS generation in human CD68lowCD14high macrophages is strongly suppressed by inhibition of dynamin. Furthermore, Nox2-deficient mice are protected against high-fat diet-induced hepatic steatosis and insulin resistance. Therefore, endocytosis of TLR4 and NOX2 into macrophages might be a novel therapeutic target for non-alcoholic fatty liver disease. Reactive species of oxygen promote the development of hepatic steatosis. Here, Kim et al. demonstrate that palmitate stimulates macrophage infiltration and increases oxidative stress during steatosis by binding to the TLR4–MD2 complex, which results in the activation of NOX2.
Collapse
|
95
|
Tucureanu MM, Rebleanu D, Constantinescu CA, Deleanu M, Voicu G, Butoi E, Calin M, Manduteanu I. Lipopolysaccharide-induced inflammation in monocytes/macrophages is blocked by liposomal delivery of G i-protein inhibitor. Int J Nanomedicine 2017; 13:63-76. [PMID: 29317816 PMCID: PMC5743190 DOI: 10.2147/ijn.s150918] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Lipopolysaccharide (LPS) is widely recognized as a potent activator of monocytes/macrophages, and its effects include an altered production of key mediators, such as inflammatory cytokines and chemokines. The involvement of Gi protein in mediating LPS effects has been demonstrated in murine macrophages and various cell types of human origin. Purpose The aim of the present work was to evaluate the potential of a Gi-protein inhibitor encapsulated in liposomes in reducing the inflammatory effects induced by LPS in monocytes/macrophages. Materials and methods Guanosine 5′-O-(2-thiodiphosphate) (GOT), a guanosine diphosphate analog that completely inhibits G-protein activation by guanosine triphosphate and its analogs, was encapsulated into liposomes and tested for anti-inflammatory effects in LPS-activated THP1 monocytes or THP1-derived macrophages. The viability of monocytes/macrophages after incubation with different concentrations of free GOT or liposome-encapsulated GOT was assessed by MTT assay. MAPK activation and production of IL1β, TNFα, IL6, and MCP1 were assessed in LPS-activated monocytes/macrophages in the presence or absence of free or encapsulated GOT. In addition, the effect of free or liposome-encapsulated GOT on LPS-stimulated monocyte adhesion to activated endothelium and on monocyte chemotaxis was evaluated. Results We report here that GOT-loaded liposomes inhibited activation of MAPK and blocked the production of the cytokines IL1β, TNFα, IL6, and MCP1 induced by LPS in monocytes and macrophages. Moreover, GOT encapsulated in liposomes reduced monocyte adhesion and chemotaxis. All demonstrated events were in contrast with free GOT, which showed reduced or no effect on monocyte/macrophage activation with LPS. Conclusion This study demonstrates the potential of liposomal GOT in blocking LPS proinflammatory effects in monocytes/macrophages.
Collapse
Affiliation(s)
- Monica Madalina Tucureanu
- Department of Biopathology and Therapy of Inflammation, Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| | - Daniela Rebleanu
- Department of Biopathology and Therapy of Inflammation, Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| | - Cristina Ana Constantinescu
- Department of Biopathology and Therapy of Inflammation, Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania.,Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, Bucharest, Romania
| | - Mariana Deleanu
- Department of Lipidomics, Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania.,Faculty of Biotechnologies, University of Agronomic Sciences and Veterinary Medicine, Bucharest, Romania
| | - Geanina Voicu
- Department of Biopathology and Therapy of Inflammation, Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| | - Elena Butoi
- Department of Biopathology and Therapy of Inflammation, Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| | - Manuela Calin
- Department of Biopathology and Therapy of Inflammation, Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| | - Ileana Manduteanu
- Department of Biopathology and Therapy of Inflammation, Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| |
Collapse
|
96
|
Acinar injury and early cytokine response in human acute biliary pancreatitis. Sci Rep 2017; 7:15276. [PMID: 29127325 PMCID: PMC5681596 DOI: 10.1038/s41598-017-15479-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 10/27/2017] [Indexed: 12/16/2022] Open
Abstract
Clinical acute pancreatitis (AP) is marked by an early phase of systemic inflammatory response syndrome (SIRS) with multiorgan dysfunction (MODS), and a late phase characterized by sepsis with MODS. However, the mechanisms of acinar injury in human AP and the associated systemic inflammation are not clearly understood. This study, for the first time, evaluated the early interactions of bile acid induced human pancreatic acinar injury and the resulting cytokine response. We exposed freshly procured resected human pancreata to taurolithocolic acid (TLCS) and evaluated for acinar injury, cytokine release and interaction with peripheral blood mononuclear cells (PBMCs). We observed autophagy in acinar cells in response to TLCS exposure. There was also time-dependent release of IL-6, IL-8 and TNF-α from the injured acini that resulted in activation of PBMCs. We also observed that cytokines secreted by activated PBMCs resulted in acinar cell apoptosis and further cytokine release from them. Our data suggests that the earliest immune response in human AP originates within the acinar cell itself, which subsequently activates circulating PBMCs leading to SIRS. These findings need further detailed evaluation so that specific therapeutic targets to curb SIRS and resulting early adverse outcomes could be identified and tested.
Collapse
|
97
|
Redox regulation in tumor cell epithelial-mesenchymal transition: molecular basis and therapeutic strategy. Signal Transduct Target Ther 2017; 2:17036. [PMID: 29263924 PMCID: PMC5661624 DOI: 10.1038/sigtrans.2017.36] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 04/25/2017] [Accepted: 04/25/2017] [Indexed: 02/05/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is recognized as a driving force of cancer cell metastasis and drug resistance, two leading causes of cancer recurrence and cancer-related death. It is, therefore, logical in cancer therapy to target the EMT switch to prevent such cancer metastasis and recurrence. Previous reports have indicated that growth factors (such as epidermal growth factor and fibroblast growth factor) and cytokines (such as the transforming growth factor beta (TGF-β) family) are major stimulators of EMT. However, the mechanisms underlying EMT initiation and progression remain unclear. Recently, emerging evidence has suggested that reactive oxygen species (ROS), important cellular secondary messengers involved in diverse biological events in cancer cells, play essential roles in the EMT process in cancer cells by regulating extracellular matrix (ECM) remodeling, cytoskeleton remodeling, cell–cell junctions, and cell mobility. Thus, targeting EMT by manipulating the intracellular redox status may hold promise for cancer therapy. Herein, we will address recent advances in redox biology involved in the EMT process in cancer cells, which will contribute to the development of novel therapeutic strategies by targeting redox-regulated EMT for cancer treatment.
Collapse
|
98
|
Hwang D, Jo H, Kim JK, Lim YH. Oxyresveratrol-containing Ramulus mori ethanol extract attenuates acute colitis by suppressing inflammation and increasing mucin secretion. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.05.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
99
|
Solution structure of the TLR adaptor MAL/TIRAP reveals an intact BB loop and supports MAL Cys91 glutathionylation for signaling. Proc Natl Acad Sci U S A 2017; 114:E6480-E6489. [PMID: 28739909 DOI: 10.1073/pnas.1701868114] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MyD88 adaptor-like (MAL) is a critical protein in innate immunity, involved in signaling by several Toll-like receptors (TLRs), key pattern recognition receptors (PRRs). Crystal structures of MAL revealed a nontypical Toll/interleukin-1 receptor (TIR)-domain fold stabilized by two disulfide bridges. We therefore undertook a structural and functional analysis of the role of reactive cysteine residues in the protein. Under reducing conditions, the cysteines do not form disulfides, but under oxidizing conditions they are highly amenable to modification. The solution structure of the reduced form of the MAL TIR domain, determined by NMR spectroscopy, reveals a remarkable structural rearrangement compared with the disulfide-bonded structure, which includes the relocation of a β-strand and repositioning of the functionally important "BB-loop" region to a location more typical for TIR domains. Redox measurements by NMR further reveal that C91 has the highest redox potential of all cysteines in MAL. Indeed, mass spectrometry revealed that C91 undergoes glutathionylation in macrophages activated with the TLR4 ligand lipopolysaccharide (LPS). The C91A mutation limits MAL glutathionylation and acts as a dominant negative, blocking the interaction of MAL with its downstream target MyD88. The H92P mutation mimics the dominant-negative effects of the C91A mutation, presumably by preventing C91 glutathionylation. The MAL C91A and H92P mutants also display diminished degradation and interaction with interleukin-1 receptor-associated kinase 4 (IRAK4). We conclude that in the cell, MAL is not disulfide-bonded and requires glutathionylation of C91 for signaling.
Collapse
|
100
|
Ji M, Park CK, Lee JW, Park KY, Son KH, Hong JH. Two Phase Modulation of [Formula: see text] Entry and Cl -/[Formula: see text] Exchanger in Submandibular Glands Cells by Dexmedetomidine. Front Physiol 2017; 8:86. [PMID: 28298895 PMCID: PMC5331071 DOI: 10.3389/fphys.2017.00086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 01/30/2017] [Indexed: 12/15/2022] Open
Abstract
Dexmedetomidine (Dex), a highly selective α2-adrenoceptor agonist, attenuates inflammatory responses induced by lipopolysaccharide (LPS) and induces sedative and analgesic effects. Administration of Dex also reduces salivary secretion in human subjects and inhibits osmotic water permeability in rat cortical collecting ducts. However, little is known about the mechanisms underlying the effects of Dex on salivary glands fluid secretion. We demonstrated the α2-adrenoceptor expression in the basolateral membrane of mouse submandibular glands (SMG). To investigate fluid secretion upon treatment with Dex, we studied the effects of Dex on the activity of Na+-K+-2Cl- cotransporter1 (NKCC1) and Cl-/[Formula: see text] exchange (CBE), and on downstream pro-inflammatory cytokine expression in isolated primary mouse SMG cells. Dex acutely increased CBE activity and NKCC1-mediated and independent [Formula: see text] entry in SMG duct cells, and enhanced ductal fluid secretion in a sealed duct system. Dex showed differential effects on cholinergic/adrenergic stimulations and inflammatory mediators, histamine, and LPS, stimulations-induced Ca2+ in mouse SMG cells. Both, histamine- and LPS-induced intracellular Ca2+ increases were inhibited by Dex, whereas carbachol-stimulated Ca2+ signals were not. Long-lasting (2 h) treatment with Dex reduced CBE activity in SMG and in human submandibular glands (HSG) cells. Moreover, when isolated SMG cells were stimulated with Dex for 2 h, phosphodiesterase 4D (PDE4D) expression was enhanced. These results confirm the anti-inflammatory properties of Dex on LPS-mediated signaling. Further, Dex also inhibited mRNA expression of interleukin-6 and NADPH oxidase 4. The present study also showed that α2-adrenoceptor activation by Dex reduces salivary glands fluid secretion by increasing PDE4D expression, and subsequently reducing the concentration of cAMP. These findings reveal an interaction between the α2-adrenoceptor and PDE4D, which should be considered when using α2-adrenoceptor agonists as sedative or analgesics.
Collapse
Affiliation(s)
- Minjeong Ji
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon UniversityIncheon, South Korea
| | - Chul-Kyu Park
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon UniversityIncheon, South Korea
| | - Jin Woo Lee
- Department of Molecular Medicine, School of Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon UniversityIncheon, South Korea
| | - Kook Yang Park
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon UniversityIncheon, South Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon UniversityIncheon, South Korea
| | - Jeong Hee Hong
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon UniversityIncheon, South Korea
| |
Collapse
|