1
|
Cerdeira CD, Brigagão MRPL. Targeting Macrophage Polarization in Infectious Diseases: M1/M2 Functional Profiles, Immune Signaling and Microbial Virulence Factors. Immunol Invest 2024; 53:1030-1091. [PMID: 38913937 DOI: 10.1080/08820139.2024.2367682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
INTRODUCTION An event of increasing interest during host-pathogen interactions is the polarization of patrolling/naive monocytes (MOs) into macrophage subsets (MФs). Therapeutic strategies aimed at modulating this event are under investigation. METHODS This review focuses on the mechanisms of induction/development and profile of MФs polarized toward classically proinflammatory (M1) or alternatively anti-inflammatory (M2) phenotypes in response to bacteria, fungi, parasites, and viruses. RESULTS AND DISCUSSION It highlights nuclear, cytoplasmic, and cell surface receptors (pattern recognition receptors/PPRs), microenvironmental mediators, and immune signaling. MФs polarize into phenotypes: M1 MФs, activated by IFN-γ, pathogen-associated molecular patterns (PAMPs, e.g. lipopolysaccharide) and membrane-bound PPRs ligands (TLRs/CLRs ligands); or M2 MФs, induced by interleukins (ILs-4, -10 and -13), antigen-antibody complexes, and helminth PAMPs. Polarization toward M1 and M2 profiles evolve in a pathogen-specific manner, with or without canonicity, and can vary widely. Ultimately, this can result in varying degrees of host protection or more severe disease outcome. On the one hand, the host is driving effective MФs polarization (M1 or M2); but on the other hand, microorganisms may skew the polarization through virulence factors to increase pathogenicity. Cellular/genomic reprogramming also ensures plasticity of M1/M2 phenotypes. Because modulation of polarization can occur at multiple points, new insights and emerging perspectives may have clinical implications during the inflammation-to-resolution transition; translated into practical applications as for therapeutic/vaccine design target to boost microbicidal response (M1, e.g. triggering oxidative burst) with specifics PAMPs/IFN-γ or promote tissue repair (M2, increasing arginase activity) via immunotherapy.
Collapse
|
2
|
Pan S, Peng L, Yi Q, Qi W, Yang H, Wang H, Wang L. Ginsenoside Rh 2 Alleviates LPS-Induced Inflammatory Responses by Binding to TLR 4/MD-2 and Blocking TLR 4 Dimerization. Int J Mol Sci 2024; 25:9546. [PMID: 39273493 PMCID: PMC11395244 DOI: 10.3390/ijms25179546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Lipopolysaccharide (LPS) triggers a severe systemic inflammatory reaction in mammals, with the dimerization of TLR4/MD-2 upon LPS stimulation serving as the pivotal mechanism in the transmission of inflammatory signals. Ginsenoside Rh2 (G-Rh2), one of the active constituents of red ginseng, exerts potent anti-inflammatory activity. However, whether G-Rh2 can block the TLR4 dimerization to exert anti-inflammatory effects remains unclear. Here, we first investigated the non-cytotoxic concentration of G-Rh2 on RAW 264.7 cells, and detected the releases of pro-inflammatory cytokines in LPS-treated RAW 264.7 cells, and then uncovered the mechanisms involved in the anti-inflammatory activity of G-Rh2 through flow cytometry, fluorescent membrane localization, Western blotting, co-immunoprecipitation (Co-IP), molecular docking and surface plasmon resonance (SPR) analysis in LPS-stimulated macrophages. Our results show that G-Rh2 stimulation markedly inhibited the secretion of LPS-induced interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and nitric oxide (NO). Additionally, G-Rh2 blocked the binding of LPS with the membrane of RAW 264.7 cells through direct interaction with TLR4 and MD-2 proteins, leading to the disruption of the dimerization of TLR4 and MD-2, followed by suppression of the TLR4/NF-κB signaling pathway. Our results suggest that G-Rh2 acts as a new inhibitor of TLR4 dimerization and may serve as a promising therapeutic agent against inflammation.
Collapse
Affiliation(s)
- Shujuan Pan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China; (S.P.); (H.W.)
- Engineering Research Center of the Utilization for Characteristic Bio-Pharma Ceutical Resources in Southwest, Ministry of Education, Guizhou University, Guiyang 550025, China; (Q.Y.); (W.Q.); (H.Y.)
| | - Luyuan Peng
- College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| | - Qion Yi
- Engineering Research Center of the Utilization for Characteristic Bio-Pharma Ceutical Resources in Southwest, Ministry of Education, Guizhou University, Guiyang 550025, China; (Q.Y.); (W.Q.); (H.Y.)
| | - Weijin Qi
- Engineering Research Center of the Utilization for Characteristic Bio-Pharma Ceutical Resources in Southwest, Ministry of Education, Guizhou University, Guiyang 550025, China; (Q.Y.); (W.Q.); (H.Y.)
| | - Hui Yang
- Engineering Research Center of the Utilization for Characteristic Bio-Pharma Ceutical Resources in Southwest, Ministry of Education, Guizhou University, Guiyang 550025, China; (Q.Y.); (W.Q.); (H.Y.)
| | - Hongying Wang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China; (S.P.); (H.W.)
- Engineering Research Center of the Utilization for Characteristic Bio-Pharma Ceutical Resources in Southwest, Ministry of Education, Guizhou University, Guiyang 550025, China; (Q.Y.); (W.Q.); (H.Y.)
| | - Lu Wang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China; (S.P.); (H.W.)
- Engineering Research Center of the Utilization for Characteristic Bio-Pharma Ceutical Resources in Southwest, Ministry of Education, Guizhou University, Guiyang 550025, China; (Q.Y.); (W.Q.); (H.Y.)
| |
Collapse
|
3
|
Zhang S, Liu Y, Zhang XL, Sun Y, Lu ZH. ANKRD22 aggravates sepsis-induced ARDS and promotes pulmonary M1 macrophage polarization. J Transl Autoimmun 2024; 8:100228. [PMID: 38225946 PMCID: PMC10788270 DOI: 10.1016/j.jtauto.2023.100228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 01/17/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is independently associated with a poor prognosis in patients with sepsis. Macrophage M1 polarization plays an instrumental role in this process. Therefore, the exploration of key molecules affecting acute lung injury and macrophage M1 polarization may provide therapeutic targets for the treatment of septic ARDS. Here, we identified that elevated levels of Ankyrin repeat domain-containing protein 22 (ANKRD22) were associated with poor prognosis and more pronounced M1 macrophage polarization in septic patients by analyzing high-throughput data. ANKRD22 expression was also significantly upregulated in the alveolar lavage fluid, peripheral blood, and lung tissue of septic ARDS model mice. Knockdown of ANKRD22 significantly attenuated acute lung injury in mice with sepsis-induced ARDS and reduced the M1 polarization of lung macrophages. Furthermore, deletion of ANKRD22 in macrophages inhibited M1 macrophage polarization and reduced levels of phosphorylated IRF3 and intracellular interferon regulatory factor 3 (IRF3) expression, while re-expression of ANKRD22 reversed these changes. Further experiments revealed that ANKRD22 promotes IRF3 activation by binding to mitochondrial antiviral-signaling protein (MAVS). In conclusion, these findings suggest that ANKRD22 promotes the M1 polarization of lung macrophages and exacerbates sepsis-induced ARDS.
Collapse
Affiliation(s)
- Shi Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, ZhongdaHospital, Southeast University, Nanjing, Jiangsu, China
- Department of Pulmonary and Critical Care Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yao Liu
- Emergency Department of Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Gulou District, Nanjing, China
| | - Xiao-Long Zhang
- Department of Ultrasound, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yun Sun
- The First Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province, 230601, China
| | - Zhong-Hua Lu
- The First Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province, 230601, China
| |
Collapse
|
4
|
Santos TCD, Obando JMC, Leite PEC, Pereira MR, Leitão MDF, Abujadi C, Pimenta LDFL, Martins RCC, Cavalcanti DN. Approaches of marine compounds and relevant immune mediators in Autism Spectrum Disorder: Opportunities and challenges. Eur J Med Chem 2024; 266:116153. [PMID: 38277916 DOI: 10.1016/j.ejmech.2024.116153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/12/2024] [Accepted: 01/13/2024] [Indexed: 01/28/2024]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder that affects social skills, language, communication, and behavioral skills, significantly impacting the individual's quality of life. Recently, numerous works have centered on the connections between the immune and central nervous systems and the influence of neuroinflammation on autism symptomatology. Marine natural products are considered as important alternative sources of different types of compounds, including polysaccharides, polyphenols, sterols, carotenoids, terpenoids and, alkaloids. These compounds present anti-inflammatory, neuroprotective and immunomodulatory activities, exhibiting a potential for the treatment of many diseases. Although many studies address the marine compounds in the modulation of inflammatory mediators, there is a gap regarding their use in the regulation of the immune system in ASD. Thus, this review aims to provide a better understanding regarding cytokines, chemokines, growth factors and immune responses in ASD, as well as the potential of bioactive marine compounds in the immune regulation in ASD. We expect that this review would contribute to the development of therapeutic alternatives for controlling immune mediators and inflammation in ASD.
Collapse
Affiliation(s)
- Thalisia Cunha Dos Santos
- Programa de Pós-graduação em Química de Produtos Naturais, Instituto de Pesquisas de Produtos Naturais Walter Mors, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Laboratório de Produtos Naturais de Algas Marinha (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - Johana Marcela Concha Obando
- Laboratório de Produtos Naturais de Algas Marinha (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Paulo Emílio Corrêa Leite
- Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Programa de Pós-graduação em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Instituto LisMAPS, Niterói, RJ, Brazil
| | - Mariana Rodrigues Pereira
- Programa de Pós-graduação em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Instituto LisMAPS, Niterói, RJ, Brazil; Programa de Pós-graduação em Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Mônica de Freitas Leitão
- Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Faculdade de Medicina, Pontifícia Universidade Católica de Campinas (PUC-Camp), Campinas, SP, Brazil
| | - Caio Abujadi
- Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Programa de Pós-graduação em Ciência, Tecnologia e Inclusão (PGCTIn), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | | | - Roberto Carlos Campos Martins
- Programa de Pós-graduação em Química de Produtos Naturais, Instituto de Pesquisas de Produtos Naturais Walter Mors, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diana Negrão Cavalcanti
- Laboratório de Produtos Naturais de Algas Marinha (ALGAMAR), Departamento de Biologia Marinha, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Núcleo de Estudos e Pesquisas em Autismo (NEPA), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil; Programa de Pós-graduação em Ciência, Tecnologia e Inclusão (PGCTIn), Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| |
Collapse
|
5
|
Hu C. Marine natural products and human immunity: novel biomedical resources for anti-infection of SARS-CoV-2 and related cardiovascular disease. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:12. [PMID: 38282092 PMCID: PMC10822835 DOI: 10.1007/s13659-024-00432-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/17/2024] [Indexed: 01/30/2024]
Abstract
Marine natural products (MNPs) and marine organisms include sea urchin, sea squirts or ascidians, sea cucumbers, sea snake, sponge, soft coral, marine algae, and microalgae. As vital biomedical resources for the discovery of marine drugs, bioactive molecules, and agents, these MNPs have bioactive potentials of antioxidant, anti-infection, anti-inflammatory, anticoagulant, anti-diabetic effects, cancer treatment, and improvement of human immunity. This article reviews the role of MNPs on anti-infection of coronavirus, SARS-CoV-2 and its major variants (such as Delta and Omicron) as well as tuberculosis, H. Pylori, and HIV infection, and as promising biomedical resources for infection related cardiovascular disease (irCVD), diabetes, and cancer. The anti-inflammatory mechanisms of current MNPs against SARS-CoV-2 infection are also discussed. Since the use of other chemical agents for COVID-19 treatment are associated with some adverse effects in cardiovascular system, MNPs have more therapeutic advantages. Herein, it's time to protect this ecosystem for better sustainable development in the new era of ocean economy. As huge, novel and promising biomedical resources for anti-infection of SARS-CoV-2 and irCVD, the novel potential mechanisms of MNPs may be through multiple targets and pathways regulating human immunity and inhibiting inflammation. In conclusion, MNPs are worthy of translational research for further clinical application.
Collapse
Affiliation(s)
- Chunsong Hu
- Department of Cardiovascular Medicine, Jiangxi Academy of Medical Science, Nanchang University, Hospital of Nanchang University, No. 461 Bayi Ave, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
6
|
Su J, Zhou F, Wu S, Tong Z. Research Progress on Natural Small-Molecule Compounds for the Prevention and Treatment of Sepsis. Int J Mol Sci 2023; 24:12732. [PMID: 37628912 PMCID: PMC10454676 DOI: 10.3390/ijms241612732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Sepsis is a serious disease with high mortality and has been a hot research topic in medical research in recent years. With the continuous reporting of in-depth research on the pathological mechanisms of sepsis, various compounds have been developed to prevent and treat sepsis. Natural small-molecule compounds play vital roles in the prevention and treatment of sepsis; for example, compounds such as resveratrol, emodin, salidroside, ginsenoside, and others can modulate signaling through the NF-κB, STAT3, STAT1, PI3K, and other pathways to relieve the inflammatory response, immunosuppression, and organ failure caused by sepsis. Here, we discuss the functions and mechanisms of natural small-molecule compounds in preventing and treating sepsis. This review will lay the theoretical foundation for discovering new natural small-molecule compounds that can potentially prevent and treat sepsis.
Collapse
|
7
|
Mojiri-Forushani H, Khajehali E, Adelipour M, Mohammadi A. Inhibitory effects of fluoxetine on the secretion of inflammatory mediators and JAK/STAT3 and JNK/TLR4 gene expression. Mol Biol Rep 2023; 50:2231-2241. [PMID: 36571654 PMCID: PMC9791631 DOI: 10.1007/s11033-022-08219-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/16/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Selective serotonin reuptake inhibitors (SSRIs) are the most common class of medicines used for the treatment of major depression. Recent studies have reported an association between depression and inflammation and suggested the significant effects of SSRIs on inflammatory processes. METHODS The current study aimed to evaluate the effects of fluoxetine, an SSRI, on the level of inflammatory cytokines, including interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), in the rat serum and RAW264.7 mouse macrophage cell line, using ELISA sandwich assays. Also, the expression of inflammatory genes, including JAK/STAT3 and TLR4/JNK, was examined in macrophages, using real-time quantitative reverse transcription PCR to determine the potential mechanism of fluoxetine in inflammation. The rats received fluoxetine (10, 20, and 40 mg/kg) 30 min before lipopolysaccharide (LPS) treatment for 90 min. The cells received different doses of fluoxetine (5, 10, and 20 µg/mL) before stimulation with LPS for 24 or 48 h. RESULTS The serum concentrations of IL-1β, IL-6, and TNF-α were reduced in rats and cells treated with fluoxetine. Following fluoxetine administration, the expression of JAK/STAT3 and TLR4/JNK genes was significantly decreased in the RAW264.7 cells treated with LPS for 24 h. However, after 48 h of treatment with LPS, fluoxetine failed to diminish the elevated expression of JAK and JNK genes, while it significantly decreased the expression of STAT3 and TLR4 genes. CONCLUSION The findings revealed that fluoxetine has anti-inflammatory properties, mainly due to the reduction of inflammatory cytokines and inhibition of JAK/STAT3 and TLR4/JNK gene expression in macrophages.
Collapse
Affiliation(s)
| | - Elham Khajehali
- Department of Anatomy & Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Victoria, 3010, Australia.
| | - Maryam Adelipour
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
8
|
Deficiency of mindin reduces renal injury after ischemia reperfusion. Mol Med 2022; 28:152. [PMID: 36510147 PMCID: PMC9743537 DOI: 10.1186/s10020-022-00578-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Acute renal injury (AKI) secondary to ischemia reperfusion (IR) injury continues to be a significant perioperative problem and there is no effective treatment. Mindin belongs to the mindin/F-spondin family and involves in inflammation, proliferation, and cell apoptosis. Previous studies have explored the biological functions of mindin in liver and brain ischemic injury, but its role in AKI is unknown. METHOD To investigate whether mindin has a pathogenic role, mindin knockout (KO) and wild-type (WT) mice were used to establish renal IR model. After 30 min of ischemia and 24 h of reperfusion, renal histology, serum creatinine, and inflammatory response were examined to assess kidney injury. In vitro, proinflammatory factors and inflammatory signaling pathways were measured in mindin overexpression or knockdown and vector cells after hypoxia/reoxygenation (HR). RESULTS Following IR, the kidney mindin level was increased in WT mice and deletion of mindin provided significant protection for mice against IR-induced renal injury as manifested by attenuated the elevation of serum creatinine and blood urea nitrogen along with less severity for histological alterations. Mindin deficiency significantly suppressed inflammatory cell infiltration, TNF-α and MCP-1 production following renal IR injury. Mechanistic studies revealed that mindin deficiency inhibits TLR4/JNK/NF-κB signaling activation. In vitro, the expression levels of TNF-α and MCP-1 were increased in mindin overexpression cells compared with vector cells following HR. Moreover, TLR4/JNK/NF-κB signaling activation was elevated in the mindin overexpression cells in response to HR stimulation while mindin knockdown inhibited the activation of TLR4/JNK/ NF-κB signaling after HR in vitro. Further study showed that mindin protein interacted directly with TLR4 protein. And more, mindin protein was confirmed to be expressed massively in renal tubule tissues of human hydronephrosis patients. CONCLUSION These data demonstrate that mindin is a critical modulator of renal IR injury through regulating inflammatory responses. TLR4/JNK/NF-κB signaling most likely mediates the biological function of mindin in this model of renal ischemia.
Collapse
|
9
|
Chen Z, Lin H, Han J, Fang D, Wang M, Liao J. Enantioselective Copper-Catalyzed Electrophilic Sulfenylation of Cyclic Imino Esters. Org Lett 2021; 23:9146-9150. [PMID: 34787420 DOI: 10.1021/acs.orglett.1c03464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Herein we report an enantioselective sulfenylation of cyclic imino esters with the efficient and versatile sulfenylation reagent S-alkyl 4-methylbenzenesulfonothioates. By utilizing the Cu/tBu-Phosferrox catalytic system, we can assemble diverse S-alkyl groups into the cyclic imino esters under mild conditions in good yields and with excellent enantioselectivities. Remarkably, this method demonstrates a high tolerance of diverse functional groups and proves to be applicable in the late-stage functionalization of pharmaceuticals.
Collapse
Affiliation(s)
- Zhiwei Chen
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huaxin Lin
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Han
- College of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Dongmei Fang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Min Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Liao
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,College of Chemical Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
10
|
Margalef J, Samec JSM. Assessing Methodologies to Synthesize α-Sulfenylated Carbonyl Compounds by Green Chemistry Metrics. CHEMSUSCHEM 2021; 14:808-823. [PMID: 33180999 PMCID: PMC7894555 DOI: 10.1002/cssc.202002409] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/08/2020] [Indexed: 06/11/2023]
Abstract
α-Sulfenylated carbonyl compounds are important both as active pharmaceutical ingredients and as intermediates in organic synthesis. Owing to their relevance in synthetic organic chemistry, this Minireview focuses on assessing the most relevant synthetic procedures based on green chemistry metrics. The Minireview starts with the traditional routes and then focuses on more recently developed methodologies. These routes include sulfenylating reagents using organocatalysis, cross-dehydrogenative couplings using in situ halogenations to prevent reactive intermediates in high concentrations, oxidative couplings using terminal oxidants such as DDQ or TEMPO, and redox-neutral couplings using transition metal catalysis. These methodologies have been evaluated on the basis of atom economy, E factor, and the safety and toxicity of the transformations and the solvents used. Besides using green metrics to evaluate these novel methodologies, the synthetic utility is also assessed with regard to the availability of starting materials and the generality of the reactions. This Minireview aims to inspire researchers to apply green assessments to other methodologies and also for them to take measures to increase the greenness of their developed transformations.
Collapse
Affiliation(s)
- Jèssica Margalef
- Departament de Química Física i InorgànicaUniversitat Rovira i VirgiliC/ Marcel lí Domingo, 143007TarragonaSpain
| | - Joseph S. M. Samec
- Department of Organic ChemistryStockholm UniversitySvante Arrhenius väg 16 C106 91StockholmSweden
| |
Collapse
|
11
|
Chen H, Jiang W, Zeng Q. Recent Advances in Synthesis of Chiral Thioethers. CHEM REC 2020; 20:1269-1296. [PMID: 32930488 DOI: 10.1002/tcr.202000084] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022]
Abstract
Chiral thioethers is an important class of organosulfur molecules with extensive applications, especially in the field of medicine and organic synthesis. This review discusses the recent progress of synthesis of enantioenriched chiral thioethers and hopes to be helpful for related research in the future. It is summarized from organosulfur compounds-participating organic reaction types, including nucleophilic substitution, cross coupling, sulfa-Michael addition, sulfenylation, asymmetric allylic reaction, asymmetric Doyle-Kirmse reaction, Pummerer-type rearrangement, Smiles rearrangement,[2,3] Stevens and Sommelet-Hauser rearrangement.
Collapse
Affiliation(s)
- Hongyi Chen
- State Key Laboratory of Geohazard Prevention and Geoenvironment Protection, College of Materials, Chemistry & Chemical Engineering, Chengdu University of Technology, 1 Dongsan Road, Erxianqiao, Chengdu, 610059, China
| | - Wenlong Jiang
- State Key Laboratory of Geohazard Prevention and Geoenvironment Protection, College of Materials, Chemistry & Chemical Engineering, Chengdu University of Technology, 1 Dongsan Road, Erxianqiao, Chengdu, 610059, China
| | - Qingle Zeng
- State Key Laboratory of Geohazard Prevention and Geoenvironment Protection, College of Materials, Chemistry & Chemical Engineering, Chengdu University of Technology, 1 Dongsan Road, Erxianqiao, Chengdu, 610059, China
| |
Collapse
|
12
|
Occurrence and Properties of Thiosilvatins. Mar Drugs 2019; 17:md17120664. [PMID: 31779089 PMCID: PMC6950259 DOI: 10.3390/md17120664] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 12/02/2022] Open
Abstract
The spread of studies on biodiversity in different environmental contexts is particularly fruitful for natural product discovery, with the finding of novel secondary metabolites and structural models, which are sometimes specific to certain organisms. Within the large class of the epipolythiodioxopiperazines, which are typical of fungi, thiosilvatins represent a homogeneous family that, so far, has been reported in low frequency in both marine and terrestrial contexts. However, recent observations indicate that these compounds have been possibly neglected in the metabolomic characterization of fungi, particularly from marine sources. Aspects concerning occurrence, bioactivities, structural, and biosynthetic properties of thiosilvatins are reviewed in this paper.
Collapse
|
13
|
Hepatoprotective effect of ultrasonicated ginseng berry extract on a rat mild bile duct ligation model. J Ginseng Res 2019; 43:606-617. [PMID: 31695567 PMCID: PMC6823758 DOI: 10.1016/j.jgr.2018.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/27/2018] [Accepted: 07/17/2018] [Indexed: 12/28/2022] Open
Abstract
Background The Panax ginseng berry extract (GBE) is well known to have an antidiabetic effect. The aim of this study is to evaluate and investigate the protective effect of ultrasonication-processed P. ginseng berry extract (UGBE) compared with GBE on liver fibrosis induced by mild bile duct ligation (MBDL) model in rats. After ultrasonication process, the composition ratio of ginsenoside in GBE was changed. The component ratio of ginsenosides Rh1, Rh4, Rg2, Rg3, Rk1, Rk3, and F4 in the extract was elevated. Methods In this study, the protective effect of the newly developed UGBE was evaluated on hepatotoxicity and neuronal damage in MBDL model. Silymarin (150 mg/kg) was used for positive control. UGBE (100 mg/kg, 250 mg/kg, 500 mg/kg), GBE (250 mg/kg), and silymarin (150 mg/kg) were orally administered for 6 weeks after MBDL surgery. Results The MBDL surgery induced severe hepatotoxicity that leads to liver inflammation in rats. Also, the serum ammonia level was increased by MBDL surgery. However, the liver dysfunction of MBDL surgery–operated rats was attenuated by UGBE treatment via myeloid differentiation factor 88-dependent Toll-like receptor 4 signaling pathways. Conclusion UGBE has a protective effect on liver fibrosis induced by MBDL in rats through inhibition of the TLR4 signaling pathway in liver.
Collapse
|
14
|
Saqib U, Sarkar S, Suk K, Mohammad O, Baig MS, Savai R. Phytochemicals as modulators of M1-M2 macrophages in inflammation. Oncotarget 2018; 9:17937-17950. [PMID: 29707159 PMCID: PMC5915167 DOI: 10.18632/oncotarget.24788] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/25/2018] [Indexed: 02/07/2023] Open
Abstract
Macrophages are critical mediators of the innate immune response against foreign pathogens, including bacteria, physical stress, and injury. Therefore, these cells play a key role in the "inflammatory pathway" which in turn can lead to an array of diseases and disorders such as autoimmune neuropathies and myocarditis, inflammatory bowel disease, atherosclerosis, sepsis, arthritis, diabetes, and angiogenesis. Recently, more studies have focused on the macrophages inflammatory diseases since the discovery of the two subtypes of macrophages, which are differentiated on the basis of their phenotype and distinct gene expression pattern. Of these, M1 macrophages are pro-inflammatory and responsible for inflammatory signaling, while M2 are anti-inflammatory macrophages that participate in the resolution of the inflammatory process, M2 macrophages produce anti-inflammatory cytokines, thereby contributing to tissue healing. Many studies have shown the role of these two subtypes in the inflammatory pathway, and their emergence appears to decide the fate of inflammatory signaling and disease progression. As a next step in directing the pro-inflammatory response toward the anti-inflammatory type after an insult by a foreign pathogen (e. g., bacterial lipopolysaccharide), investigators have identified many natural compounds that have the potential to modulate M1 to M2 macrophages. In this review, we provide a focused discussion of advances in the identification of natural therapeutic molecules with anti-inflammatory properties that modulate the phenotype of macrophages from M1 to M2.
Collapse
Affiliation(s)
- Uzma Saqib
- Discipline of Chemistry, School of Basic Sciences, Indian Institute of Technology (IIT) Indore, MP, India
| | - Sutripta Sarkar
- PostGraduate Department of Food & Nutrition, BRSN College (affiliated to WBSU), Kolkata, WB, India
| | - Kyoungho Suk
- Department of Pharmacology, Kyungpook National University School of Medicine, Joong-gu Daegu, South Korea
| | - Owais Mohammad
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University (AMU), Aligarh, UP, India
| | - Mirza S Baig
- Discipline of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology (IIT), Indore, MP, India
| | - Rajkumar Savai
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen 35392, Germany.,Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Member of the DZL, Bad Nauheim, Germany
| |
Collapse
|
15
|
Zhao Y, Zhu H, Wang H, Ding L, Xu L, Chen D, Shen S, Hou Y, Dou H. FC-99 ameliorates sepsis-induced liver dysfunction by modulating monocyte/macrophage differentiation via Let-7a related monocytes apoptosis. Oncotarget 2018; 9:14959-14976. [PMID: 29599918 PMCID: PMC5871089 DOI: 10.18632/oncotarget.24127] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 12/03/2017] [Indexed: 12/14/2022] Open
Abstract
Background The liver is a vital target for sepsis-related injury, leading to inflammatory pathogenesis, multiple organ dysfunction and high mortality rates. Monocyte-derived macrophage transformations are key events in hepatic inflammation. N1-[(4-methoxy)methyl]-4-methyl-1,2-benzenediamine (FC-99) previously displayed therapeutic potential on experimental sepsis. However, the underlying mechanism of this protective effect is still not clear. Results FC-99 treatment attenuated the liver dysfunction in septic mice that was accompanied with reduced numbers of pro-inflammatory Ly6Chi monocytes in the peripheral blood and CD11b+F4/80lo monocyte-derived macrophages in the liver. These effects were attributed to the FC-99-induced apoptosis of CD11b+ cells. In PMA-differentiated THP-1 cells, FC-99 repressed the expression of CD11b, CD14 and caspase3 and resulted in a high proportion of Annexin V+ cells. Moreover, let-7a-5p expression was abrogated upon CLP stimulation in vivo, whereas it was restored by FC-99 treatment. TargetScan analysis and luciferase assays indicated that the anti-apoptotic protein BCL-XL was targeted by let-7a-5p. BCL-XL was inhibited by FC-99 in order to induce monocyte apoptosis, leading to the impaired monocyte-to-macrophage differentiation. Materials and Methods Murine acute liver failure was generated by caecal ligation puncture surgery after FC-99 administration; Blood samples and liver tissues were collected to determine the monocyte/macrophage subsets and the induction of apoptosis. Human acute monocytic leukemia cell line (THP-1) cells were pretreated with FC-99 followed by phorbol-12-myristate-13-acetate (PMA) stimulation, in order to induce monocyte-to-macrophage differentiation. The target of FC-99 and the mechanistic analyses were conducted by microarrays, qRT-PCR validation, TargetScan algorithms and a luciferase report assay. Conclusions FC-99 exhibits potential therapeutic effects on CLP-induced liver dysfunction by restoring let-7a-5p levels.
Collapse
Affiliation(s)
- Yarong Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, PR China
| | - Haiyan Zhu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, PR China
| | - Haining Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, PR China
| | - Liang Ding
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, PR China
| | - Lizhi Xu
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, PR China
| | - Dai Chen
- Novel Bioinformatics Co., Ltd, Shanghai, PR China
| | - Sunan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, PR China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, PR China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, PR China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, PR China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, PR China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, PR China
| |
Collapse
|
16
|
Wang X, Li Y, Zhang X, Lai D, Zhou L. Structural Diversity and Biological Activities of the Cyclodipeptides from Fungi. Molecules 2017; 22:E2026. [PMID: 29168781 PMCID: PMC6149763 DOI: 10.3390/molecules22122026] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 11/15/2017] [Indexed: 11/17/2022] Open
Abstract
Cyclodipeptides, called 2,5-diketopiperazines (2,5-DKPs), are obtained by the condensation of two amino acids. Fungi have been considered to be a rich source of novel and bioactive cyclodipeptides. This review highlights the occurrence, structures and biological activities of the fungal cyclodipeptides with the literature covered up to July 2017. A total of 635 fungal cyclodipeptides belonging to the groups of tryptophan-proline, tryptophan-tryptophan, tryptophan-Xaa, proline-Xaa, non-tryptophan-non-proline, and thio-analogs have been discussed and reviewed. They were mainly isolated from the genera of Aspergillus and Penicillium. More and more cyclodipeptides have been isolated from marine-derived and plant endophytic fungi. Some of them were screened to have cytotoxic, phytotoxic, antimicrobial, insecticidal, vasodilator, radical scavenging, antioxidant, brine shrimp lethal, antiviral, nematicidal, antituberculosis, and enzyme-inhibitory activities to show their potential applications in agriculture, medicinal, and food industry.
Collapse
Affiliation(s)
- Xiaohan Wang
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Yuying Li
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Xuping Zhang
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Daowan Lai
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Ligang Zhou
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
17
|
TLR4 Signaling Pathway Modulators as Potential Therapeutics in Inflammation and Sepsis. Vaccines (Basel) 2017; 5:vaccines5040034. [PMID: 28976923 PMCID: PMC5748601 DOI: 10.3390/vaccines5040034] [Citation(s) in RCA: 367] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/29/2017] [Accepted: 10/01/2017] [Indexed: 02/06/2023] Open
Abstract
Toll-Like Receptor 4 (TLR4) signal pathway plays an important role in initiating the innate immune response and its activation by bacterial endotoxin is responsible for chronic and acute inflammatory disorders that are becoming more and more frequent in developed countries. Modulation of the TLR4 pathway is a potential strategy to specifically target these pathologies. Among the diseases caused by TLR4 abnormal activation by bacterial endotoxin, sepsis is the most dangerous one because it is a life-threatening acute system inflammatory condition that still lacks specific pharmacological treatment. Here, we review molecules at a preclinical or clinical phase of development, that are active in inhibiting the TLR4-MyD88 and TLR4-TRIF pathways in animal models. These are low-molecular weight compounds of natural and synthetic origin that can be considered leads for drug development. The results of in vivo studies in the sepsis model and the mechanisms of action of drug leads are presented and critically discussed, evidencing the differences in treatment results from rodents to humans.
Collapse
|
18
|
Mayer AMS, Rodríguez AD, Taglialatela-Scafati O, Fusetani N. Marine Pharmacology in 2012-2013: Marine Compounds with Antibacterial, Antidiabetic, Antifungal, Anti-Inflammatory, Antiprotozoal, Antituberculosis, and Antiviral Activities; Affecting the Immune and Nervous Systems, and Other Miscellaneous Mechanisms of Action. Mar Drugs 2017; 15:md15090273. [PMID: 28850074 PMCID: PMC5618412 DOI: 10.3390/md15090273] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/17/2017] [Accepted: 08/21/2017] [Indexed: 12/23/2022] Open
Abstract
The peer-reviewed marine pharmacology literature from 2012 to 2013 was systematically reviewed, consistent with the 1998–2011 reviews of this series. Marine pharmacology research from 2012 to 2013, conducted by scientists from 42 countries in addition to the United States, reported findings on the preclinical pharmacology of 257 marine compounds. The preclinical pharmacology of compounds isolated from marine organisms revealed antibacterial, antifungal, antiprotozoal, antituberculosis, antiviral and anthelmitic pharmacological activities for 113 marine natural products. In addition, 75 marine compounds were reported to have antidiabetic and anti-inflammatory activities and affect the immune and nervous system. Finally, 69 marine compounds were shown to display miscellaneous mechanisms of action which could contribute to novel pharmacological classes. Thus, in 2012–2013, the preclinical marine natural product pharmacology pipeline provided novel pharmacology and lead compounds to the clinical marine pharmaceutical pipeline, and contributed significantly to potentially novel therapeutic approaches to several global disease categories.
Collapse
Affiliation(s)
- Alejandro M S Mayer
- Department of Pharmacology, Chicago College of Osteopathic Medicine, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA.
| | - Abimael D Rodríguez
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce de León Avenue, San Juan, PR 00926, USA.
| | | | | |
Collapse
|
19
|
Thomas GW, Rael LT, Mains CW, Slone D, Carrick MM, Bar-Or R, Bar-Or D. Anti-Inflammatory Activity in the Low Molecular Weight Fraction of Commercial Human Serum Albumin (LMWF5A). J Immunoassay Immunochem 2016; 37:55-67. [PMID: 25961642 DOI: 10.1080/15321819.2015.1047516] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The innate immune system is increasingly being recognized as a critical component in osteoarthritis (OA) pathophysiology. An ex vivo immunoassay utilizing human peripheral blood mononuclear cells (PBMC) was developed in order to assess the OA anti-inflammatory properties of the low molecular weight fraction (<5 kDa) of commercial human serum albumin (LMWF5A). PBMC from various donors were pre-incubated with LMWF5A before LPS stimulation. TNFα release was measured by ELISA in supernatants after an overnight incubation. A ≥ 30% decrease in TNFα release was observed. This anti-inflammatory effect is potentially useful in assessing potency of LMWF5A for the treatment of OA.
Collapse
Affiliation(s)
- Gregory W Thomas
- a Swedish Medical Center , Trauma Research Department , Englewood , Colorado , USA.,b St. Anthony Hospital , Lakewood , Colorado , USA.,c The Medical Center of Plano , Plano , Texas , USA
| | - Leonard T Rael
- a Swedish Medical Center , Trauma Research Department , Englewood , Colorado , USA.,b St. Anthony Hospital , Lakewood , Colorado , USA.,c The Medical Center of Plano , Plano , Texas , USA
| | - Charles W Mains
- b St. Anthony Hospital , Lakewood , Colorado , USA.,d Rocky Vista University , Parker , Colorado , USA
| | - Denetta Slone
- a Swedish Medical Center , Trauma Research Department , Englewood , Colorado , USA.,d Rocky Vista University , Parker , Colorado , USA
| | | | - Raphael Bar-Or
- a Swedish Medical Center , Trauma Research Department , Englewood , Colorado , USA.,b St. Anthony Hospital , Lakewood , Colorado , USA.,c The Medical Center of Plano , Plano , Texas , USA
| | - David Bar-Or
- a Swedish Medical Center , Trauma Research Department , Englewood , Colorado , USA.,b St. Anthony Hospital , Lakewood , Colorado , USA.,c The Medical Center of Plano , Plano , Texas , USA.,d Rocky Vista University , Parker , Colorado , USA
| |
Collapse
|
20
|
Feng X, Yu W, Zhou F, Chen J, Shen P. A novel small molecule compound diaporine inhibits breast cancer cell proliferation via promoting ROS generation. Biomed Pharmacother 2016; 83:1038-1047. [DOI: 10.1016/j.biopha.2016.08.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/01/2016] [Accepted: 08/09/2016] [Indexed: 11/26/2022] Open
|
21
|
Yu JS, Huang HM, Ding PG, Hu XS, Zhou F, Zhou J. Catalytic Enantioselective Construction of Sulfur-Containing Tetrasubstituted Carbon Stereocenters. ACS Catal 2016. [DOI: 10.1021/acscatal.6b01496] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jin-Sheng Yu
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes, School of
Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, P. R. China
| | - Hong-Mei Huang
- College
of Chemistry and Material Sciences, Sichuan Normal University, Chengdu, Sichuan 610066, P. R. China
| | - Pei-Gang Ding
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes, School of
Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, P. R. China
| | - Xiao-Si Hu
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes, School of
Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, P. R. China
| | - Feng Zhou
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes, School of
Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, P. R. China
| | - Jian Zhou
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes, School of
Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, P. R. China
- State
Key Laboratory of Organometallic Chemistry, Shanghai Institute of
Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
| |
Collapse
|
22
|
Abstract
This review covers the literature published in 2013 for marine natural products (MNPs), with 982 citations (644 for the period January to December 2013) referring to compounds isolated from marine microorganisms and phytoplankton, green, brown and red algae, sponges, cnidarians, bryozoans, molluscs, tunicates, echinoderms, mangroves and other intertidal plants and microorganisms. The emphasis is on new compounds (1163 for 2013), together with the relevant biological activities, source organisms and country of origin. Reviews, biosynthetic studies, first syntheses, and syntheses that lead to the revision of structures or stereochemistries, have been included.
Collapse
Affiliation(s)
- John W Blunt
- Department of Chemistry, University of Canterbury, Christchurch, New Zealand.
| | | | | | | | | |
Collapse
|
23
|
Metabolic profile of 2-(2-hydroxypropanamido) benzoic acid in rats by ultra high performance liquid chromatography combined with Fourier transform ion cyclotron resonance mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 993-994:60-8. [DOI: 10.1016/j.jchromb.2015.04.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/19/2015] [Accepted: 04/25/2015] [Indexed: 11/23/2022]
|
24
|
LI FANGFANG, YUAN YUAN, LIU YUAN, WU QINGQING, JIAO RONG, YANG ZHENG, ZHOU MENGQIAO, TANG QIZHU. Pachymic acid protects H9c2 cardiomyocytes from lipopolysaccharide-induced inflammation and apoptosis by inhibiting the extracellular signal-regulated kinase 1/2 and p38 pathways. Mol Med Rep 2015; 12:2807-13. [DOI: 10.3892/mmr.2015.3712] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 02/06/2015] [Indexed: 11/05/2022] Open
|
25
|
Kouwenhoven EN, van Bokhoven H, Zhou H. Gene regulatory mechanisms orchestrated by p63 in epithelial development and related disorders. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:590-600. [PMID: 25797018 DOI: 10.1016/j.bbagrm.2015.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/09/2015] [Accepted: 03/12/2015] [Indexed: 01/30/2023]
Abstract
The transcription factor p63 belongs to the p53 family and is a key regulator in epithelial commitment and development. Mutations in p63 give rise to several epithelial related disorders with defects in skin, limb and orofacial structures. Since the discovery of p63, efforts have been made to identify its target genes using individual gene approaches and to understand p63 function in normal epithelial development and related diseases. Recent genome-wide approaches have identified tens of thousands of potential p63-regulated target genes and regulatory elements, and reshaped the concept of gene regulation orchestrated by p63. These data also provide insights into p63-related disease mechanisms. In this review, we discuss the regulatory role of p63 in normal and diseased epithelial development in light of these novel findings. We also propose future perspectives for dissecting the molecular mechanism of p63-mediated epithelial development and related disorders as well as for potential therapeutic strategies.
Collapse
Affiliation(s)
- Evelyn N Kouwenhoven
- Radboud University, Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.
| | - Hans van Bokhoven
- Radboud university medical center, Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.
| | - Huiqing Zhou
- Radboud University, Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands; Radboud university medical center, Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Li X, Huang S, Ren Y, Wang M, Kang C, Xie L, Shi D. Establishment of a mouse model to express bovine CD14 short hairpin RNA. BMC Vet Res 2015; 11:36. [PMID: 25889660 PMCID: PMC4332730 DOI: 10.1186/s12917-015-0353-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 02/06/2015] [Indexed: 11/25/2022] Open
Abstract
Background Cluster of differentiation 14 (CD14) functions as a co-receptor for Toll-like receptor (TLR)-4 and myeloid differentiation factor (MD)-2 in detecting bacterial lipopolysaccharide. Together, these complexes promote the phagocytosis and digestion of Gram-negative bacteria, and initiate immune responses. To date, much of our understanding of CD14 function during Gram-negative bacterial inflammation comes from studies on mouse knockout models and cell transfection. To identify the effect of CD14 knockdown in this process in large livestock animals, we established a mouse model expressing bovine CD14 short hairpin (sh) RNA. shRNA fragments targeting bovine CD14 were screened by co-transfection in HEK 293 cells, and the most effective CD14 shRNA fragment was cloned into the eukaryotic expression vector pSilencer4.1-CD14 shRNA-IRES (internal ribosome entry site) and transferred into mouse zygotes by pronuclear microinjection to obtain transgenic mice. Expression of the enhanced green fluorescent protein (EGFP) reporter and genes related to the TLR4 signaling pathway was detected by immunohistochemistry (IHC) and quantitative polymerase chain reaction (PCR), respectively. Results One effective shRNA fragment (shRNA-674) targeting bovine CD14 was obtained, the sequence of which was shown to be conserved between cows, buffalos, sheep, and humans. Thirty-seven founder pups were obtained by pronuclear microinjection, of which three were positive for the transgene. In the F1 generation, 11 of 33 mice (33%) were positive for the transgene as detected by PCR. IHC analysis detected exogenous EGFP expression in the liver, kidney, and spleen of transgenic F1 mice, indicating that they were chimeric. The expression of endogenous CD14 mRNA in the heart, liver, spleen, lung, and kidney of transgenic F1 mice was decreased 8-, 3-, 19.5-, 6-, and 11-fold, respectively. The expression patterns of endogenous MD-2, TLR4, interleukin-6 and tumor necrosis factor-α genes in transgenic mice also varied. Conclusions This study confirms that transgenic mice expressing bovine CD14 shRNA can be generated by pronuclear microinjection, and demonstrates inhibited endogenous mouse CD14 expression that alters gene expression related to the TLR4 signaling pathway.
Collapse
Affiliation(s)
- Xiangping Li
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University, Nanning, Guangxi, China. .,Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, Guangxi University, Nanning, 530004, China.
| | - Shihai Huang
- College of Life Science and Technology, Guangxi University, Nanning, Guangxi, China.
| | - Yanping Ren
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University, Nanning, Guangxi, China. .,Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, Guangxi University, Nanning, 530004, China.
| | - Meng Wang
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University, Nanning, Guangxi, China. .,Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, Guangxi University, Nanning, 530004, China.
| | - Chao Kang
- College of Life Science and Technology, Guangxi University, Nanning, Guangxi, China.
| | - Liangliang Xie
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University, Nanning, Guangxi, China. .,Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, Guangxi University, Nanning, 530004, China.
| | - Deshun Shi
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University, Nanning, Guangxi, China. .,Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, Guangxi University, Nanning, 530004, China.
| |
Collapse
|
27
|
Li X, Li M, Huang S, Qiao S, Qin Z, Kang C, Shi D. The effect of buffalo CD14 shRNA on the gene expression of TLR4 signal pathway in buffalo monocyte/macrophages. Cell Mol Biol Lett 2014; 19:623-37. [PMID: 25355240 PMCID: PMC6275898 DOI: 10.2478/s11658-014-0217-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/24/2014] [Indexed: 11/25/2022] Open
Abstract
CD14 plays a crucial role in the inflammatory response to lipopolysaccharide (LPS), which interacts with TLR4 and MD-2 to enable cell activation, resulting in inflammation. Upstream inhibition of the inflammation pathway mediated by bacterial LPS, toll-like receptor 4 (TLR4) and cluster of differentiation antigen 14 (CD14) was proven to be an effective therapeutic approach for attenuating harmful immune activation. To explore the effect of CD14 downregulation on the expression of TLR4 signaling pathway-related genes after LPS stimulation in buffalo (Bubalus bubalis) monocyte/macrophages, effective CD14 shRNA sequences were screened using qRT-PCR and FACS analysis with buffalo CD14 shRNA lentiviral recombinant plasmids (pSicoRGFP-shRNA) and buffalo CD14 fusion expression plasmids (pDsRed-N1-buffalo CD14) co-transfected into HEK293T cells via liposomes. Of the tested shRNAs, shRNA-1041 revealed the highest knockdown efficiency (p < 0.01). When buffalo peripheral blood monocyte/macrophages were infected with shRNA-1041 lentivirus and stimulated with LPS, the expression of endogenous CD14 was significantly decreased by CD14 shRNA (p < 0.01), and the mRNA expression levels of TLR4, IL-6 and TNF-α were also significantly downregulated compared to the control groups (p < 0.01). These results demonstrated that the knockdown of endogenous CD14 had clear regulatory effects on the signal transduction of TLR4 after stimulation with LPS. These results may provide a better understanding of the molecular mechanisms of CD14 regulation in the development of several buffalo diseases.
Collapse
Affiliation(s)
- Xiangping Li
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University, Nanning, Guangxi, China
| | - Meiqing Li
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University, Nanning, Guangxi, China
| | - Shihai Huang
- College of life science and technology, Guangxi University, Nanning, Guangxi, China
| | - Shuye Qiao
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University, Nanning, Guangxi, China
| | - Zhaoxian Qin
- Guangxi Institute of Animal Science, Nanning, Guangxi, China
| | - Chao Kang
- College of life science and technology, Guangxi University, Nanning, Guangxi, China
| | - Deshun Shi
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University, Nanning, Guangxi, China
| |
Collapse
|
28
|
Shukla P, Rao GM, Pandey G, Sharma S, Mittapelly N, Shegokar R, Mishra PR. Therapeutic interventions in sepsis: current and anticipated pharmacological agents. Br J Pharmacol 2014; 171:5011-31. [PMID: 24977655 PMCID: PMC4253453 DOI: 10.1111/bph.12829] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/29/2014] [Accepted: 06/13/2014] [Indexed: 12/14/2022] Open
Abstract
Sepsis is a clinical syndrome characterized by a multisystem response to a pathogenic assault due to underlying infection that involves a combination of interconnected biochemical, cellular and organ-organ interactive networks. After the withdrawal of recombinant human-activated protein C (rAPC), researchers and physicians have continued to search for new therapeutic approaches and targets against sepsis, effective in both hypo- and hyperinflammatory states. Currently, statins are being evaluated as a viable option in clinical trials. Many agents that have shown favourable results in experimental sepsis are not clinically effective or have not been clinically evaluated. Apart from developing new therapeutic molecules, there is great scope for for developing a variety of drug delivery strategies, such as nanoparticulate carriers and phospholipid-based systems. These nanoparticulate carriers neutralize intracorporeal LPS as well as deliver therapeutic agents to targeted tissues and subcellular locations. Here, we review and critically discuss the present status and new experimental and clinical approaches for therapeutic intervention in sepsis.
Collapse
Affiliation(s)
- Prashant Shukla
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - G Madhava Rao
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - Gitu Pandey
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - Shweta Sharma
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - Naresh Mittapelly
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| | - Ranjita Shegokar
- Department of Pharmaceutics, Biopharmaceutics & NutriCosmetics, Institute of Pharmacy, Freie Universität BerlinBerlin, Germany
| | - Prabhat Ranjan Mishra
- Pharmaceutics Division, Preclinical South PCS 002/011, CSIR – Central Drug Research InstituteLucknow, India
| |
Collapse
|
29
|
Vitamin C mitigates oxidative stress and tumor necrosis factor-alpha in severe community-acquired pneumonia and LPS-induced macrophages. Mediators Inflamm 2014; 2014:426740. [PMID: 25253919 PMCID: PMC4165740 DOI: 10.1155/2014/426740] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/16/2014] [Indexed: 01/09/2023] Open
Abstract
Oxidative stress is an important part of host innate immune response to foreign pathogens. However, the impact of vitamin C on oxidative stress and inflammation remains unclear in community-acquired pneumonia (CAP). We aimed to determine the effect of vitamin C on oxidative stress and inflammation. CAP patients were enrolled. Reactive oxygen species (ROS), DNA damage, superoxide dismutases (SOD) activity, tumor necrosis factor-alpha (TNF-α), and IL-6 were analyzed in CAP patients and LPS-stimulated macrophages cells. MH-S cells were transfected with RFP-LC3 plasmids. Autophagy was measured in LPS-stimulated macrophages cells. Severe CAP patients showed significantly increased ROS, DNA damage, TNF-α, and IL-6. SOD was significantly decreased in severe CAP. Vitamin C significantly decreased ROS, DNA damage, TNF-α, and IL-6. Vitamin C inhibited LPS-induced ROS, DNA damage, TNF-α, IL-6, and p38 in macrophages cells. Vitamin C inhibited autophagy in LPS-induced macrophages cells. These findings indicated that severe CAP exhibited significantly increased oxidative stress, DNA damage, and proinflammatory mediator. Vitamin C mitigated oxidative stress and proinflammatory mediator suggesting a possible mechanism for vitamin C in severe CAP.
Collapse
|
30
|
Song Y, Dou H, Wang P, Zhao S, Wang T, Gong W, Zhao J, Li E, Tan R, Hou Y. A novel small-molecule compound diaporine A inhibits non-small cell lung cancer growth by regulating miR-99a/mTOR signaling. Cancer Biol Ther 2014; 15:1423-30. [PMID: 25046358 DOI: 10.4161/cbt.29925] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs (miRNAs) dysregulation is critically involved in lung cancer. Regulating miRNAs by natural agents may be a new strategy for cancer treatment. We previously found that a novel small-molecule compound diaporine A (D261), a natural product of endophytic fungus 3lp-10, had potential anti-cancer activites. In the present study, the inhibitory effect of D261 on non-small cell lung cancer (NSCLC) growth and its possible mechanisms involving miRNA regulation were investigated. By cell viability assay, cell proliferation analysis, and clonal growth assay, we proved that D261 effectively inhibited the proliferation of NSCLC cells (NCI-H460 and A549) in vitro. Administration of D261 (5 mg/kg) to NCI-H460 xenografts bearing mice also inhibited tumor growth and decreased the expression of cell proliferation regulator, midkine. Moreover, D261 induced cell cycle arrest with a reduced expression of various G 1/S transition-related molecules including cyclin D1, cyclin E1, CDK4, and CDK2, but without influencing apoptosis in NSCLC cells. Intriguingly, D261 modified expressions of some miRNAs and especially upregulated miR-99a, whose direct target was mammalian target of rapamycin (mTOR). Furthermore, overexpression of miR-99a antagonized the anti-tumor actions of D261 including the suppression of mTOR pathway activation, cell cycle-related proteins and cell growth. In addition, blocking of miR-99a expression by transfection of miR-99a inhibitors before D261 treatment counteracted the anti-tumor effects of D261. These data suggest that miR-99a/mTOR pathway was involved in D261-induced tumor suppression in NSCLC cells. D261 might be a potent anti-cancer agent by upregulating miR-99a expression.
Collapse
Affiliation(s)
- Yuxian Song
- The State Key Laboratory of Pharmaceutical Biotechnology; Division of Immunology; Medical School; Nanjing University; Nanjing, PR China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology; Division of Immunology; Medical School; Nanjing University; Nanjing, PR China
| | - Ping Wang
- The State Key Laboratory of Pharmaceutical Biotechnology; Division of Immunology; Medical School; Nanjing University; Nanjing, PR China
| | - Shuli Zhao
- Central Laboratory of Nanjing First Hospital; Nanjing Medical University; Nanjing, PR China
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology; Division of Immunology; Medical School; Nanjing University; Nanjing, PR China
| | - Wei Gong
- The State Key Laboratory of Pharmaceutical Biotechnology; Division of Immunology; Medical School; Nanjing University; Nanjing, PR China
| | - Junli Zhao
- Nanjing Xiaozhuang College; Nanjing, PR China
| | - Erguang Li
- The State Key Laboratory of Pharmaceutical Biotechnology; Division of Immunology; Medical School; Nanjing University; Nanjing, PR China
| | - Renxiang Tan
- Institute of Functional Biomolecules; State Key Laboratory of Pharmaceutical Biotechnology; School of Lifesciences; Nanjing University; Nanjing, PR China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology; Division of Immunology; Medical School; Nanjing University; Nanjing, PR China; Jiangsu Key Laboratory of Molecular Medicine; Nanjing, PR China
| |
Collapse
|
31
|
Wang XT, Tu WJ, Liu LM, Liang DY, Yu FP, Zhao L, Ye CG, Yang ZW, Gao DY. Urantide inhibits hepatic IRF3 expression in acute liver failure mice. Shijie Huaren Xiaohua Zazhi 2014; 22:2559-2564. [DOI: 10.11569/wcjd.v22.i18.2559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of urantide, a urotensin Ⅱ (UⅡ) receptor inhibitor, on interferon regulatory factor 3 (IRF3) expression in the liver tissue of mice with acute liver failure (ALF).
METHODS: Male Balb/c mice were randomly divided into four groups (n = 6 for each group): normal control, pre-treatment control, model and pre-treatment model. The pre-treatment mice received urantide (0.6 mg/kg body weight) via a caudal vein injection. At 30 min post-injection, the model (including pre-treatment model) mice were treated with lipopolysaccharide (LPS)/D-galactosamine (D-GalN) to induce ALF via an intraperitoneal injection. Liver tissues were sampled 12 h after LPS/D-GalN injection. IRF3 mRNA expression was detected by RT-PCR and real-time polymerase chain reaction (PCR), and protein expression was detected by Western blot assay.
RESULTS: The relative levels of IRF3 mRNA were significantly higher in model mice than in control and pretreatment control mice (P < 0.001 for all). Compared with the model group, pretreatment model mice had significant lower levels of IRF3 mRNA (P < 0.001). IRF3 protein levels were also significantly higher in model mice than in control and pretreatment control mice (P < 0.001 for all), while the protein levels were significantly lower in pretreatment model mice than in model mice (P < 0.05).
CONCLUSION: Urantide can inhibit the up-regulation of IRF3 mRNA and protein expression in the liver tissue of mice with LPS/D-GalN-induced ALF.
Collapse
|
32
|
Labonte AC, Tosello-Trampont AC, Hahn YS. The role of macrophage polarization in infectious and inflammatory diseases. Mol Cells 2014; 37:275-85. [PMID: 24625576 PMCID: PMC4012075 DOI: 10.14348/molcells.2014.2374] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 12/16/2013] [Indexed: 02/08/2023] Open
Abstract
Macrophages, found in circulating blood as well as integrated into several tissues and organs throughout the body, represent an important first line of defense against disease and a necessary component of healthy tissue homeostasis. Additionally, macrophages that arise from the differentiation of monocytes recruited from the blood to inflamed tissues play a central role in regulating local inflammation. Studies of macrophage activation in the last decade or so have revealed that these cells adopt a staggering range of phenotypes that are finely tuned responses to a variety of different stimuli, and that the resulting subsets of activated macrophages play critical roles in both progression and resolution of disease. This review summarizes the current understanding of the contributions of differentially polarized macrophages to various infectious and inflammatory diseases and the ongoing effort to develop novel therapies that target this key aspect of macrophage biology.
Collapse
Affiliation(s)
- Adam C. Labonte
- Department of Microbiology, Beirne B. Carter Center for Immunology Research, University of Virginia,
USA
| | | | - Young S. Hahn
- Department of Microbiology, Beirne B. Carter Center for Immunology Research, University of Virginia,
USA
| |
Collapse
|
33
|
Stonik VA, Fedorov SN. Marine low molecular weight natural products as potential cancer preventive compounds. Mar Drugs 2014; 12:636-71. [PMID: 24473167 PMCID: PMC3944507 DOI: 10.3390/md12020636] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/14/2014] [Accepted: 01/15/2014] [Indexed: 12/17/2022] Open
Abstract
Due to taxonomic positions and special living environments, marine organisms produce secondary metabolites that possess unique structures and biological activities. This review is devoted to recently isolated and/or earlier described marine compounds with potential or established cancer preventive activities, their biological sources, molecular mechanisms of their action, and their associations with human health and nutrition. The review covers literature published in 2003–2013 years and focuses on findings of the last 2 years.
Collapse
Affiliation(s)
- Valentin A Stonik
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Prospect 100 let Vladivostoku, 159, Vladivostok 690950, Russia.
| | - Sergey N Fedorov
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Prospect 100 let Vladivostoku, 159, Vladivostok 690950, Russia.
| |
Collapse
|
34
|
Qiao B, Liu X, Duan S, Yan L, Jiang Z. Highly Enantioselective Organocatalytic α-Sulfenylation of Azlactones. Org Lett 2014; 16:672-5. [PMID: 24432722 DOI: 10.1021/ol403303k] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Baokun Qiao
- Institute of Chemical Biology and ‡Key Laboratory of Natural Medicine
and Immuno-Engineering
of Henan Province, Henan University, Kaifeng, Henan, China, 475004
| | - Xinfei Liu
- Institute of Chemical Biology and ‡Key Laboratory of Natural Medicine
and Immuno-Engineering
of Henan Province, Henan University, Kaifeng, Henan, China, 475004
| | - Shaobo Duan
- Institute of Chemical Biology and ‡Key Laboratory of Natural Medicine
and Immuno-Engineering
of Henan Province, Henan University, Kaifeng, Henan, China, 475004
| | - Lin Yan
- Institute of Chemical Biology and ‡Key Laboratory of Natural Medicine
and Immuno-Engineering
of Henan Province, Henan University, Kaifeng, Henan, China, 475004
| | - Zhiyong Jiang
- Institute of Chemical Biology and ‡Key Laboratory of Natural Medicine
and Immuno-Engineering
of Henan Province, Henan University, Kaifeng, Henan, China, 475004
| |
Collapse
|
35
|
Wang Y. Attenuation of berberine on lipopolysaccharide-induced inflammatory and apoptosis responses in β-cells via TLR4-independent JNK/NF-κB pathway. PHARMACEUTICAL BIOLOGY 2013; 52:532-538. [PMID: 24188583 DOI: 10.3109/13880209.2013.840851] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Abstract Context: Toll-like receptor 4 (TLR4)-independent inflammatory and apoptosis responses contribute to β-cell failure in diabetes mellitus (DM). Berberine (BBR), a bioactive isoquinoline derivative alkaloid, ameliorates the inflammatory response in DM. Objective: This study explored the protective mechanisms of BBR on TLR4-independent inflammation response in β cells. Materials and methods: Lipopolysaccharide (LPS; 100 ng/ml) was used to induce the inflammatory response in NIT-1 and rat insulinoma (INS-1) cells for 24 h. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and colony formation assays were used for the determination of cell viability. The levels of monocyte chemoattractant protein (MCP-1), interleukin 6 (IL-6), tumor necrosis factor α (TNF-α) and insulin in cultured supernatant were detected by enzyme-linked immunosorbent assay kits. Western blot analysis was performed for the expression of p-c-Jun N-terminal kinase (JNK) and p65 NF-κB in NIT-1 cells, and p65 NF-κB in INS-1 cells. Results: BBR (1.25, 2.5 and 5 μM) or TLR4 inhibitor (TAK-242, 1 μM) increased remarkably NIT-1 cell viability by 72.6 ± 5.0, 85.9 ± 9.3, 94.7 ± 7.1 and 92.6 ± 8.4%. The EC50 of BBR was 1.14 μM. Colony formation assay showed that BBR increased the number of colonies of NIT-1 and INS-1 cells. BBR, TAK-242 or SP-600125 (1 μM) could significantly reduce the levels of MCP-1, IL-6 and TNF-α, insulin and JNK and NF-κB phosphorylation in NIT-1 cells, as well as the p65 NF-κB in INS-1 cells. Discussion and conclusion: BBR could ameliorate LPS-induced β-cell injury through the TLR4-independent JNK/NF-κB pathway. Thus, this pathway may be a potential target for the prevention and treatment of DM.
Collapse
Affiliation(s)
- Ying Wang
- Department of Endocrinology, Tianjin Pingjin Hospital , Tianjin , P.R. China
| |
Collapse
|
36
|
Lipopolysaccharide induces endoplasmic store Ca2+-dependent inflammatory responses in lung microvessels. PLoS One 2013; 8:e63465. [PMID: 23675486 PMCID: PMC3651233 DOI: 10.1371/journal.pone.0063465] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Accepted: 04/03/2013] [Indexed: 01/06/2023] Open
Abstract
The pulmonary microvasculature plays a critical role in endotoxin-induced acute lung injury. However, the relevant signaling remain unclear. Specifically the role of endothelial Ca2+ in the induction of endotoxin-mediated responses in lung microvessels remains undefined. Toward elucidating this, we used the isolated blood-perfused rat lung preparation. We loaded microvessels with the Ca2+ indicator, Fura 2 AM and then determined Ca2+ responses to infusions of lipopolysaccharide (LPS) into the microvessels. LPS induced a more than two-fold increase in the amplitude of cytosolic Ca2+ oscillations. Inhibiting inositol 1,4,5 trisphosphate receptors on endoplasmic reticulum (ER) Ca2+ stores with Xestospongin C (XeC), blocked the LPS-induced increase in the Ca2+ oscillation amplitude. However, XeC did not affect entry of external Ca2+ via plasma membrane Ca2+ channels in lung microvascular endothelial cells. This suggested that LPS augmented the oscillations via release of Ca2+ from ER stores. In addition, XeC also blocked LPS-mediated activation and nuclear translocation of nuclear factor-kappa B in lung microvessels. Further, inhibiting ER Ca2+ release blunted increases in intercellular adhesion molecule-1 expression and retention of naïve leukocytes in LPS-treated microvessels. Taken together, the data suggest that LPS-mediated Ca2+ release from ER stores underlies nuclear factor-kappa B activation and downstream inflammatory signaling in lung microvessels. Thus, we show for the first time a role for inositol 1,4,5 trisphosphate-mediated ER Ca2+ release in the induction of LPS responses in pulmonary microvascular endothelium. Mechanisms that blunt this signaling may mitigate endotoxin-induced morbidity.
Collapse
|