1
|
Taylor B, Ofori KF, Parsaeimehr A, Akdemir Evrendilek G, Attarwala T, Ozbay G. Exploring the Complexities of Seafood: From Benefits to Contaminants. Foods 2025; 14:1461. [PMID: 40361542 PMCID: PMC12071223 DOI: 10.3390/foods14091461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Seafood plays a vital role in human diets worldwide, serving as an important source of high-quality protein, omega-3 fatty acids, and essential vitamins and minerals that promote health and prevent various chronic conditions. The health benefits of seafood consumption are well documented, including a reduced risk of cardiovascular diseases, improved cognitive function, and anti-inflammatory effects. However, the safety of seafood is compromised by multiple hazards that can pose significant health risks. Pathogenic microorganisms, including bacteria, viruses, and parasites, in addition to microbial metabolites, are prominent causes of the foodborne diseases linked to seafood consumption, necessitating reliable detection and monitoring systems. Molecular biology and digital techniques have emerged as essential tools for the rapid and accurate identification of these foodborne pathogens, enhancing seafood safety protocols. Additionally, the presence of chemical contaminants such as heavy metals (e.g., mercury and lead), microplastics, and per- and polyfluoroalkyl substances (PFASs) in seafood is of increasing concern due to their potential to accumulate in the food chain and adversely affect human health. The biogenic amines formed during the microbial degradation of the proteins and allergens present in certain seafood species also contribute to food safety challenges. This review aims to address the nutritional value and health-promoting effects of seafood while exploring the multifaceted risks associated with microbial contamination, chemical pollutants, and naturally occurring substances. Emphasis is placed on enhanced surveillance, seafood traceability, sustainable aquaculture practices, and regulatory harmonization as effective strategies for controlling the risks associated with seafood consumption and thereby contributing to a safer seafood supply chain.
Collapse
Affiliation(s)
- Bettina Taylor
- Human Ecology Department, Delaware State University, Dover, DE 19901, USA;
| | - Kelvin Fynn Ofori
- Integrative PhD Program in Agriculture, Food and Environmental Sciences, College of Agriculture, Science and Technology, Delaware State University, Dover, DE 19901, USA;
| | - Ali Parsaeimehr
- Department of Agriculture and Natural Resources, Delaware State University, Dover, DE 19901, USA; (A.P.); (T.A.)
| | | | - Tahera Attarwala
- Department of Agriculture and Natural Resources, Delaware State University, Dover, DE 19901, USA; (A.P.); (T.A.)
| | - Gulnihal Ozbay
- Human Ecology Department, Delaware State University, Dover, DE 19901, USA;
- Integrative PhD Program in Agriculture, Food and Environmental Sciences, College of Agriculture, Science and Technology, Delaware State University, Dover, DE 19901, USA;
- Department of Agriculture and Natural Resources, Delaware State University, Dover, DE 19901, USA; (A.P.); (T.A.)
| |
Collapse
|
2
|
Qian H, Zhang L, Wang C, Zhang M, Wen L, Zhao W. Chlorpheniramine maleate exerts an anti-keloid activity by regulation of IL-6/JAK1/STAT3 signaling pathway. Int Immunopharmacol 2025; 148:114181. [PMID: 39879833 DOI: 10.1016/j.intimp.2025.114181] [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: 11/27/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Keloids are abnormal scars formed due to fibroblast dysfunction and excessively deposited extracellular matrix (ECM). Despite the unclear process leading to the occurrence of keloids, several studies have demonstrated that histamine and its H1 receptor can effectively regulate fibroblast functions, contributing to keloid formation. Chlorpheniramine maleate (CPM) as a first-generation H1 antihistamine has been widely applied in symptomatic treatment of allergic conditions but its effects on keloids are unknown. This study holds the objective of exploring its effect on keloids. Cell Counting Kit-8 (CCK-8), apoptosis, cell cycle and migration assays were conducted to determine the effects of CPM on human keloid fibroblasts (KFs), with its therapeutic effect evaluated by constructing a keloid model in nude mice. RNA sequencing analysis, ELISA, western blotting and immunohistochemistry assisted in examining the anti-keloid mechanism of CPM. It was observed that CPM inhibited the proliferation and migration of KFs, promoted apoptosis of KFs, and blocked the G0/G1 phase of KFs. Moreover, local intralesional injection of CPM into nude mice keloid model significantly reduced keloid scars. According to RNA sequencing analysis, the gene expression of IL-6 and JAK-STAT signaling pathway were both negatively related in CPM group versus the control group. According to the in vivo and vitro experiment results, the anti-keloid activity of CPM was attributed to its inhibitory effect on the IL-6/JAK1/STAT3 signal pathway. In summary, CPM holds great potential for localized treatment of keloids.
Collapse
Affiliation(s)
- Huan Qian
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lei Zhang
- Nursing Department, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chen Wang
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengwen Zhang
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Wen
- Nursing Department, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenxia Zhao
- School of Public Health, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
3
|
Li Y, Ai S, Li Y, Ye W, Li R, Xu X, Liu Q. The role of natural products targeting macrophage polarization in sepsis-induced lung injury. Chin Med 2025; 20:19. [PMID: 39910395 PMCID: PMC11800549 DOI: 10.1186/s13020-025-01067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/17/2025] [Indexed: 02/07/2025] Open
Abstract
Sepsis-induced acute lung injury (SALI) is characterized by a dysregulated inflammatory and immune response. As a key component of the innate immune system, macrophages play a vital role in SALI, in which a macrophage phenotype imbalance caused by an increase in M1 macrophages or a decrease in M2 macrophages is common. Despite significant advances in SALI research, effective drug therapies are still lacking. Therefore, the development of new treatments for SALI is urgently needed. An increasing number of studies suggest that natural products (NPs) can alleviate SALI by modulating macrophage polarization through various targets and pathways. This review examines the regulatory mechanisms of macrophage polarization and their involvement in the progression of SALI. It highlights how NPs mitigate macrophage imbalances to alleviate SALI, focusing on key signaling pathways such as PI3K/AKT, TLR4/NF-κB, JAK/STAT, IRF, HIF, NRF2, HMGB1, TREM2, PKM2, and exosome-mediated signaling. NPs influencing macrophage polarization are classified into five groups: terpenoids, polyphenols, alkaloids, flavonoids, and others. This work provides valuable insights into the therapeutic potential of NPs in targeting macrophage polarization to treat SALI.
Collapse
Affiliation(s)
- Yake Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Beijing Institute of Chinese Medicine, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100010, China
| | - Sinan Ai
- China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yuan Li
- Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Wangyu Ye
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Rui Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Beijing Institute of Chinese Medicine, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100010, China
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100010, China.
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100010, China.
| |
Collapse
|
4
|
Tiligada E, Stefanaki C, Ennis M, Neumann D. Opportunities and challenges in the therapeutic exploitation of histamine and histamine receptor pharmacology in inflammation-driven disorders. Pharmacol Ther 2024; 263:108722. [PMID: 39306197 DOI: 10.1016/j.pharmthera.2024.108722] [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: 05/24/2024] [Revised: 07/31/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Inflammation-driven diseases encompass a wide array of pathological conditions characterised by immune system dysregulation leading to tissue damage and dysfunction. Among the myriad of mediators involved in the regulation of inflammation, histamine has emerged as a key modulatory player. Histamine elicits its actions through four rhodopsin-like G-protein-coupled receptors (GPCRs), named chronologically in order of discovery as histamine H1, H2, H3 and H4 receptors (H1-4R). The relatively low affinity H1R and H2R play pivotal roles in mediating allergic inflammation and gastric acid secretion, respectively, whereas the high affinity H3R and H4R are primarily linked to neurotransmission and immunomodulation, respectively. Importantly, however, besides the H4R, both H1R and H2R are also crucial in driving immune responses, the H2R tending to promote yet ill-defined and unexploited suppressive, protective and/or resolving processes. The modulatory action of histamine via its receptors on inflammatory cells is described in detail. The potential therapeutic value of the most recently discovered H4R in inflammatory disorders is illustrated via a selection of preclinical models. The clinical trials with antagonists of this receptor are discussed and possible reasons for their lack of success described.
Collapse
Affiliation(s)
- Ekaterini Tiligada
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Charikleia Stefanaki
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; University Research Institute of Maternal and Child Health and Precision Medicine, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Madeleine Ennis
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Detlef Neumann
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Becker L, Holtmann D. Anti-inflammatory effects of α-humulene on the release of pro-inflammatory cytokines in lipopolysaccharide-induced THP-1 cells. Cell Biochem Biophys 2024; 82:839-847. [PMID: 38388989 PMCID: PMC11344727 DOI: 10.1007/s12013-024-01235-7] [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: 01/03/2024] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
While acute inflammation is an essential physical response to harmful external influences, the transition to chronic inflammation is problematic and associated with the development and worsening of many deadly diseases. Until now, established pharmaceutical agents have had many side effects when used for long periods. In this study, a possible anti-inflammatory effect of the sesquiterpene α-humulene on lipopolysaccharide (LPS) induction was tested. Herein, human THP-1-derived macrophages were used and their pro-inflammatory interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and interleukin-1β (IL-1β) cytokine release was measured by means of enzyme-linked immunosorbent assay. A dose-dependent effect of α-humulene on IL-6 release was observed at 0.5 and 100 µM α-humulene, with a maximum IL-6 inhibition of 60% compared to the LPS reference value after the addition of 100 µM α-humulene. TNF-α as well as IL-1β cytokine concentrations were not reduced by the addition of 0.5 and 100 µM α-humulene. This study suggests that α-humulene has potential as a promising natural alternative to established pharmaceuticals for the treatment of elevated IL-6 levels and chronic inflammation in humans.
Collapse
Affiliation(s)
- Lucas Becker
- Bioprocess Intensification, Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390, Giessen, Germany
| | - Dirk Holtmann
- Bioprocess Intensification, Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390, Giessen, Germany.
- Institute of Process Engineering in Life Sciences, Karlsruhe Institute of Technology, Fritz-Haber-Weg 4, 76131, Karlsruhe, Germany.
| |
Collapse
|
6
|
Zhao D, Wang Y, Wu S, Ji X, Gong K, Zheng H, Zhu M. Research progress on the role of macrophages in acne and regulation by natural plant products. Front Immunol 2024; 15:1383263. [PMID: 38736879 PMCID: PMC11082307 DOI: 10.3389/fimmu.2024.1383263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024] Open
Abstract
Acne vulgaris is one of the most common skin diseases. The current understanding of acne primarily revolves around inflammatory responses, sebum metabolism disorders, aberrant hormone and receptor expression, colonization by Cutibacterium acnes, and abnormal keratinization of follicular sebaceous glands. Although the precise mechanism of action remains incompletely understood, it is plausible that macrophages exert an influence on these pathological features. Macrophages, as a constituent of the human innate immune system, typically manifest distinct phenotypes across various diseases. It has been observed that the polarization of macrophages toward the M1 phenotype plays a pivotal role in the pathogenesis of acne. In recent years, extensive research on acne has revealed an increasing number of natural remedies exhibiting therapeutic efficacy through the modulation of macrophage polarization. This review investigates the role of cutaneous macrophages, elucidates their potential significance in the pathogenesis of acne, a prevalent chronic inflammatory skin disorder, and explores the therapeutic mechanisms of natural plant products targeting macrophages. Despite these insights, the precise role of macrophages in the pathogenesis of acne remains poorly elucidated. Subsequent investigations in this domain will further illuminate the pathogenesis of acne and potentially offer guidance for identifying novel therapeutic targets for this condition.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yun Wang
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shuhui Wu
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiaotian Ji
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Ke Gong
- Department of Traditional Chinese Medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Huie Zheng
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Mingfang Zhu
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
7
|
Sheng X, Li C, Wang Z, Xu Y, Sun Y, Zhang W, Liu H, Wang J. Advanced applications of strontium-containing biomaterials in bone tissue engineering. Mater Today Bio 2023; 20:100636. [PMID: 37441138 PMCID: PMC10333686 DOI: 10.1016/j.mtbio.2023.100636] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 07/15/2023] Open
Abstract
Strontium (Sr) and strontium ranelate (SR) are commonly used therapeutic drugs for patients suffering from osteoporosis. Researches have showed that Sr can significantly improve the biological activity and physicochemical properties of materials in vitro and in vivo. Therefore, a large number of strontium containing biomaterials have been developed for repairing bone defects and promoting osseointegration. In this review, we provide a comprehensive overview of Sr-containing biomaterials along with the current state of their clinical use. For this purpose, the different types of biomaterials including calcium phosphate, bioactive glass, and polymers are discussed and provided future outlook on the fabrication of the next-generation multifunctional and smart biomaterials.
Collapse
Affiliation(s)
| | | | - Zhonghan Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Yu Xu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Yang Sun
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Weimin Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Jincheng Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| |
Collapse
|
8
|
Yuan Y, Zhang Q, Wu B, Huang T, Gong P, Xiang L. Oncostatin M regulates macrophages polarization in osseointegration via yes-associated protein. Int Immunopharmacol 2023; 120:110348. [PMID: 37220694 DOI: 10.1016/j.intimp.2023.110348] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 05/25/2023]
Abstract
OBJECTIVES Oncostatin M(OSM), secreted by monocytes and macrophages, has been noted to participate in bone homeostasis and macrophage polarization, which might be regulated by yes-associated protein (YAP). This study aimed to elucidate the influence and mechanisms of OSM-YAP on macrophages polarization in osseointegration. MATERIAL AND METHODS In vitro, flow cytometry, real-time PCR, and Elisa were performed to evaluate inflammatory function in bone marrow-derived macrophages (BMDMs) with OSM, siOSMR, and YAP inhibitor verteporfin (VP). In vivo, macrophage-specific YAP-deficient mice were generated to investigate the role of OSM via YAP signaling in osseointegration. RESULTS This study demonstrated that OSM could inhibit the M1 polarization, promote the M2 polarization, and induce the expression of osteogenic-related factors via VP. The conditional knock-out of YAP inhibited the osseointegration in mice, and promoted the inflammatory reaction around the implants, while OSM could restore the effect. CONCLUSIONS Our results demonstrated that OSM might play an important role in the polarization of BMDMs, and bone formation around dental and femoral implants. This effect was closely conducted by Hippo-YAP pathway. CLINICAL SIGNIFICANCE Understanding the role and mechanism of OSM in macrophage polarization around dental implants could improve comprehension of signal network of osseointegration, and it might offer a potential target of therapies to accelerate osseointegration and reduce inflammatory reactions.
Collapse
Affiliation(s)
- Ying Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 400016, China
| | - Qin Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bingfeng Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tianyu Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Sun Q, Hu S, Lou Z, Gao J. The macrophage polarization in inflammatory dermatosis and its potential drug candidates. Biomed Pharmacother 2023; 161:114469. [PMID: 37002572 DOI: 10.1016/j.biopha.2023.114469] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Inflammatory dermatosis is characterized by persistent inflammatory infiltration and hard repair of diseased skin. As a member of the human innate immune cells, macrophages usually show different phenotypes in different diseases. The macrophage phenotype (M1/M2) imbalance caused by the increase of M1 macrophages or the decrease of M2 macrophages is common in inflammatory dermatosis. In recent years, with the deepening research on inflammatory skin diseases, more and more natural medicines/traditional Chinese medicines (TCMs), represented by Shikonin and Angelica Dahurica, have shown their therapeutic effects by affecting the polarization of macrophages. This review introduced macrophage polarization in different inflammatory dermatosis, such as psoriasis. Then summarized the natural medicines/TCMs that have potential therapeutic effects so far and introduced their mechanisms of action and the proteins/signal pathways involved. We found that the TCMs with therapeutic effects listed in this review are closely related to the theory of five flavors and four properties of Chinese medicinal, and most of them are bitter, acrid and sweet. Bitter TCMs have antipyretic, anti-inflammatory and antibacterial effects, which may improve the persistent inflammation of M1 macrophage infiltration. Acrid TCMs have the effect of promoting blood circulation, while sweet TCMs have the effect of nourishing. These 2 flavors may accelerate the repair of skin lesions of inflammatory dermatosis by affecting M2 macrophages. In conclusion, we hope to provide sufficient knowledge for natural medicine research and the development of inflammatory dermatosis related to macrophage phenotype imbalance.
Collapse
Affiliation(s)
- Qingru Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, People's Republic of China
| | - Shiyu Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, People's Republic of China
| | - Zhaohuan Lou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, People's Republic of China
| | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, People's Republic of China.
| |
Collapse
|
10
|
Mießner H, Seidel J, Smith ESJ. In vitro models for investigating itch. Front Mol Neurosci 2022; 15:984126. [PMID: 36385768 PMCID: PMC9644192 DOI: 10.3389/fnmol.2022.984126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Itch (pruritus) is a sensation that drives a desire to scratch, a behavior observed in many animals. Although generally short-lasting and not causing harm, there are several pathological conditions where chronic itch is a hallmark symptom and in which prolonged scratching can induce damage. Finding medications to counteract the sensation of chronic itch has proven difficult due to the molecular complexity that involves a multitude of triggers, receptors and signaling pathways between skin, immune and nerve cells. While much has been learned about pruritus from in vivo animal models, they have limitations that corroborate the necessity for a transition to more human disease-like models. Also, reducing animal use should be encouraged in research. However, conducting human in vivo experiments can also be ethically challenging. Thus, there is a clear need for surrogate models to be used in pre-clinical investigation of the mechanisms of itch. Most in vitro models used for itch research focus on the use of known pruritogens. For this, sensory neurons and different types of skin and/or immune cells are stimulated in 2D or 3D co-culture, and factors such as neurotransmitter or cytokine release can be measured. There are however limitations of such simplistic in vitro models. For example, not all naturally occurring cell types are present and there is also no connection to the itch-sensing organ, the central nervous system (CNS). Nevertheless, in vitro models offer a chance to investigate otherwise inaccessible specific cell–cell interactions and molecular pathways. In recent years, stem cell-based approaches and human primary cells have emerged as viable alternatives to standard cell lines or animal tissue. As in vitro models have increased in their complexity, further opportunities for more elaborated means of investigating itch have been developed. In this review, we introduce the latest concepts of itch and discuss the advantages and limitations of current in vitro models, which provide valuable contributions to pruritus research and might help to meet the unmet clinical need for more refined anti-pruritic substances.
Collapse
Affiliation(s)
- Hendrik Mießner
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Judith Seidel
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Ewan St. John Smith,
| |
Collapse
|
11
|
Gao J, Sadiq FA, Zheng Y, Zhao J, He G, Sang Y. Biofilm-based delivery approaches and specific enrichment strategies of probiotics in the human gut. Gut Microbes 2022; 14:2126274. [PMID: 36175161 PMCID: PMC9542427 DOI: 10.1080/19490976.2022.2126274] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The use of probiotics has been one of the effective strategies to restructure perturbed human gut microbiota following a disease or metabolic disorder. One of the biggest challenges associated with the use of probiotic-based gut modulation strategies is to keep the probiotic cells viable and stable during the gastrointestinal transit. Biofilm-based probiotics delivery approaches have emerged as fascinating modes of probiotic delivery in which probiotics show significantly greater tolerance and biotherapeutic potential, and interestingly probiotic biofilms can be developed on food-grade surfaces too, which is ideal for the growth and proliferation of bacterial cells for incorporation into food matrices. In addition, biofilms can be further encapsulated with food-grade materials or with bacterial self-produced biofilms. This review presents a newly emerging and unprecedently discussed techniques for the safe delivery of probiotics based on biofilms and further discusses newly emerging prebiotic materials which target specific gut microbiota groups for growth and proliferation.
Collapse
Affiliation(s)
- Jie Gao
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Faizan Ahmed Sadiq
- Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Technology & Food Sciences Unit, Melle, Belgium
| | - Yixin Zheng
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Jinrong Zhao
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Guoqing He
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China,CONTACT Guoqing He College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Yaxin Sang
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China,Yaxin Sang Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
12
|
Zhao T, Chu Z, Ma J, Ouyang L. Immunomodulation Effect of Biomaterials on Bone Formation. J Funct Biomater 2022; 13:jfb13030103. [PMID: 35893471 PMCID: PMC9394331 DOI: 10.3390/jfb13030103] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Traditional bone replacement materials have been developed with the goal of directing the osteogenesis of osteoblastic cell lines toward differentiation and therefore achieving biomaterial-mediated osteogenesis, but the osteogenic effect has been disappointing. With advances in bone biology, it has been revealed that the local immune microenvironment has an important role in regulating the bone formation process. According to the bone immunology hypothesis, the immune system and the skeletal system are inextricably linked, with many cytokines and regulatory factors in common, and immune cells play an essential role in bone-related physiopathological processes. This review combines advances in bone immunology with biomaterial immunomodulatory properties to provide an overview of biomaterials-mediated immune responses to regulate bone regeneration, as well as methods to assess the bone immunomodulatory properties of bone biomaterials and how these strategies can be used for future bone tissue engineering applications.
Collapse
Affiliation(s)
- Tong Zhao
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; (T.Z.); (Z.C.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
| | - Zhuangzhuang Chu
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; (T.Z.); (Z.C.)
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
| | - Jun Ma
- Department of General Practitioners, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Correspondence: (L.O.); (J.M.); Tel.: +86-21-52039999 (L.O.); +86-21-52039999 (J.M.)
| | - Liping Ouyang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; (T.Z.); (Z.C.)
- Correspondence: (L.O.); (J.M.); Tel.: +86-21-52039999 (L.O.); +86-21-52039999 (J.M.)
| |
Collapse
|
13
|
Oktariani AF, Ramona Y, Sudaryatma PE, Dewi IAMM, Shetty K. Role of Marine Bacterial Contaminants in Histamine Formation in Seafood Products: A Review. Microorganisms 2022; 10:microorganisms10061197. [PMID: 35744715 PMCID: PMC9227395 DOI: 10.3390/microorganisms10061197] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 02/07/2023] Open
Abstract
Histamine is a toxic biogenic amine commonly found in seafood products or their derivatives. This metabolite is produced by histamine-producing bacteria (HPB) such as Proteus vulgaris, P. mirabilis, Enterobacter aerogenes, E. cloacae, Serratia fonticola, S. liquefaciens, Citrobacter freundii, C. braakii, Clostridium spp., Raoultella planticola, R. ornithinolytica, Vibrio alginolyticus, V. parahaemolyticus, V. olivaceus, Acinetobacter lowffi, Plesiomonas shigelloides, Pseudomonas putida, P. fluorescens, Aeromonas spp., Photobacterium damselae, P. phosphoreum, P. leiognathi, P. iliopiscarium, P. kishitanii, and P. aquimaris. In this review, the role of these bacteria in histamine production in fish and seafood products with consequences for human food poisoning following consumption are discussed. In addition, methods to control their activity in countering histamine production are proposed.
Collapse
Affiliation(s)
- Adnorita Fandah Oktariani
- Doctoral Study Program of Biology, Faculty of Mathematics and Natural Sciences, Udayana University, Denpasar 80361, Bali, Indonesia;
- PT. Intimas Surya, Denpasar 80222, Bali, Indonesia
| | - Yan Ramona
- Doctoral Study Program of Biology, Faculty of Mathematics and Natural Sciences, Udayana University, Denpasar 80361, Bali, Indonesia;
- Integrated Laboratory for Biosciences and Biotechnology, Udayana University, Denpasar 80361, Bali, Indonesia
- Correspondence: (Y.R.); (K.S.); Tel.: +62-85101523213 (Y.R.)
| | | | - Ida Ayu Mirah Meliana Dewi
- School of Biology, Faculty of Mathematics and Natural Sciences, Udayana University, Denpasar 80361, Bali, Indonesia;
| | - Kalidas Shetty
- Department of Plant Sciences, North Dakota State University, Fargo, ND 58102, USA
- Correspondence: (Y.R.); (K.S.); Tel.: +62-85101523213 (Y.R.)
| |
Collapse
|
14
|
Matsuo K, Lepinski A, Chavez RD, Barruet E, Pereira A, Moody TA, Ton AN, Sharma A, Hellman J, Tomoda K, Nakamura MC, Hsiao EC. ACVR1 R206H extends inflammatory responses in human induced pluripotent stem cell-derived macrophages. Bone 2021; 153:116129. [PMID: 34311122 PMCID: PMC8803261 DOI: 10.1016/j.bone.2021.116129] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 01/01/2023]
Abstract
Macrophages play crucial roles in many human disease processes. However, obtaining large numbers of primary cells for study is often difficult. We describe 2D and 3D methods for directing human induced pluripotent stem cells (hiPSCs) into macrophages (iMACs). iMACs generated in 2D culture showed functional similarities to human primary monocyte-derived M2-like macrophages, and could be successfully polarized into a M1-like phenotype. Both M1- and M2-like iMACs showed phagocytic activity and reactivity to endogenous or exogenous stimuli. In contrast, iMACs generated by a 3D culture system showed mixed M1- and M2-like functional characteristics. 2D-iMACs from patients with fibrodysplasia ossificans progressiva (FOP), an inherited disease with progressive heterotopic ossification driven by inflammation, showed prolonged inflammatory cytokine production and higher Activin A production after M1-like polarization, resulting in dampened responses to additional LPS stimulation. These results demonstrate a simple and robust way of creating hiPSC-derived M1- and M2-like macrophage lineages, while identifying macrophages as a source of Activin A that may drive heterotopic ossification in FOP.
Collapse
Affiliation(s)
- Koji Matsuo
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA; The Institute for Human Genetics, University of California, San Francisco, CA, USA; The Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Abigail Lepinski
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA; The Institute for Human Genetics, University of California, San Francisco, CA, USA; The Program in Craniofacial Biology, University of California, San Francisco, CA, USA; Division of Graduate Medical Sciences, Boston University School of Medicine, Boston, MA, USA
| | - Robert D Chavez
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA; The Institute for Human Genetics, University of California, San Francisco, CA, USA; The Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Emilie Barruet
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA; The Institute for Human Genetics, University of California, San Francisco, CA, USA; The Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Ashley Pereira
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA; The Institute for Human Genetics, University of California, San Francisco, CA, USA; The Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Tania A Moody
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA; The Institute for Human Genetics, University of California, San Francisco, CA, USA; The Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Amy N Ton
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA; The Institute for Human Genetics, University of California, San Francisco, CA, USA; The Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Aditi Sharma
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA; The Institute for Human Genetics, University of California, San Francisco, CA, USA; The Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, USA
| | - Kiichiro Tomoda
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
| | - Mary C Nakamura
- Medical Service, San Francisco Veterans Affairs Healthcare System, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA
| | - Edward C Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA; The Institute for Human Genetics, University of California, San Francisco, CA, USA; The Program in Craniofacial Biology, University of California, San Francisco, CA, USA.
| |
Collapse
|
15
|
Buelow LM, Hoji A, Tat K, Schroeder-Carter LM, Carroll DJ, Cook-Mills JM. Mechanisms for Alternaria alternata Function in the Skin During Induction of Peanut Allergy in Neonatal Mice With Skin Barrier Mutations. FRONTIERS IN ALLERGY 2021; 2:677019. [PMID: 35387035 PMCID: PMC8974772 DOI: 10.3389/falgy.2021.677019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 08/16/2021] [Indexed: 01/07/2023] Open
Abstract
Neonatal mice with heterozygous mutations in genes encoding the skin barrier proteins filaggrin and mattrin (flaky tail mice [FT+/-]) exhibit oral peanut-induced anaphylaxis after skin sensitization. As we have previously reported, sensitization in this model is achieved via skin co- exposure to the environmental allergen Alternaria alternata (Alt), peanut extract (PNE), and detergent. However, the function of Alt in initiation of peanut allergy in this model is little understood. The purpose of this study was to investigate candidate cytokines induced by Alt in the skin and determine the role of these cytokines in the development of food allergy, namely oncostatin M (Osm), amphiregulin (Areg), and IL-33. RT-qPCR analyses demonstrated that skin of FT+/- neonates expressed Il33 and Osm following Alt or Alt/PNE but not PNE exposure. By contrast, expression of Areg was induced by either Alt, PNE, or Alt/PNE sensitization in FT+/- neonates. In scRNAseq analyses, Osm, Areg, and Il33 were expressed by several cell types, including a keratinocyte cluster that was expanded in the skin of Alt/PNE-exposed FT+/- pups as compared to Alt/PNE-exposed WT pups. Areg and OSM were required for oral PNE-induced anaphylaxis since anaphylaxis was inhibited by administration of neutralizing anti-Areg or anti-OSM antibodies prior to each skin sensitization with Alt/PNE. It was then determined if intradermal injection of recombinant IL33 (rIL33), rAreg, or rOSM in the skin could substitute for Alt during skin sensitization to PNE. PNE skin sensitization with intradermal rIL33 was sufficient for oral PNE-induced anaphylaxis, whereas skin sensitization with intradermal rAreg or rOSM during skin exposure to PNE was not sufficient for anaphylaxis to oral PNE challenge. Based on these studies a pathway for IL33, Areg and OSM in Alt/PNE sensitized FT+/- skin was defined for IgE induction and anaphylaxis. Alt stimulated two pathways, an IL33 pathway and a pathway involving OSM and Areg. These two pathways acted in concert with PNE to induce food allergy in pups with skin barrier mutations.
Collapse
|
16
|
Effects of histamine and various histamine receptor antagonists on gene expression profiles of macrophages during compressive strain. J Orofac Orthop 2021; 83:13-23. [PMID: 34228141 PMCID: PMC9569297 DOI: 10.1007/s00056-021-00318-x] [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: 11/20/2020] [Accepted: 05/01/2021] [Indexed: 11/14/2022]
Abstract
Purpose Tissue hormone histamine can accumulate locally within the periodontal ligament via nutrition or may be released during allergic reactions by mast cells, which may have an impact on orthodontic tooth movement. In addition to periodontal ligament fibroblasts, cells of the immune system such as macrophages are exposed to compressive strain. The aim of this study was thus to investigate the impact of histamine on the gene expression profile of macrophages in the context of simulated orthodontic compressive strain. Methods Macrophages were incubated with different histamine concentrations (50, 100, 200 µM) for 24 h and then either left untreated or compressed for another 4 h. To assess the role of different histamine receptors, we performed experiments with antagonists for histamine 1 receptor (cetirizine), histamine 2 receptor (ranitidine) and histamine 4 receptor (JNJ7777120) under control and pressure conditions. We tested for lactate dehydrogenase release and analyzed the expression of genes involved in inflammation and bone remodeling by reverse transcription quantitative polymerase chain reaction (RT-qPCR). Results Histamine elevated gene expression of tumor necrosis factor under control conditions and in combination with pressure application. Increased prostaglandin-endoperoxide synthase‑2 mRNA was observed when histamine was combined with compressive force. Interleukin‑6 gene expression was not affected by histamine treatment. In macrophages, compressive strain increased osteoprotegerin gene expression. Histamine further elevated this effect. Most of the observed histamine effects were blocked by the histamine 1 receptor antagonist cetirizine. Conclusions Histamine has an impact on the gene expression profile of macrophages during compressive strain in vitro, most likely having an impairing effect on orthodontic tooth movement by upregulation of osteoprotegerin expression.
Collapse
|
17
|
Wong LS, Yen YT, Lee CH. The Implications of Pruritogens in the Pathogenesis of Atopic Dermatitis. Int J Mol Sci 2021; 22:7227. [PMID: 34281281 PMCID: PMC8269281 DOI: 10.3390/ijms22137227] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 01/17/2023] Open
Abstract
Atopic dermatitis (AD) is a prototypic inflammatory disease that presents with intense itching. The pathophysiology of AD is multifactorial, involving environmental factors, genetic susceptibility, skin barrier function, and immune responses. A recent understanding of pruritus transmission provides more information about the role of pruritogens in the pathogenesis of AD. There is evidence that pruritogens are not only responsible for eliciting pruritus, but also interact with immune cells and act as inflammatory mediators, which exacerbate the severity of AD. In this review, we discuss the interaction between pruritogens and inflammatory molecules and summarize the targeted therapies for AD.
Collapse
Affiliation(s)
- Lai-San Wong
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Yu-Ta Yen
- Department of Dermatology, Fooying University Hospital, Pingtung 928, Taiwan;
| | - Chih-Hung Lee
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| |
Collapse
|
18
|
The Function of the Histamine H4 Receptor in Inflammatory and Inflammation-Associated Diseases of the Gut. Int J Mol Sci 2021; 22:ijms22116116. [PMID: 34204101 PMCID: PMC8200986 DOI: 10.3390/ijms22116116] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Histamine is a pleiotropic mediator involved in a broad spectrum of (patho)-physiological processes, one of which is the regulation of inflammation. Compounds acting on three out of the four known histamine receptors are approved for clinical use. These approved compounds comprise histamine H1-receptor (H1R) antagonists, which are used to control allergic inflammation, antagonists at H2R, which therapeutically decrease gastric acid release, and an antagonist at H3R, which is indicated to treat narcolepsy. Ligands at H4R are still being tested pre-clinically and in clinical trials of inflammatory diseases, including rheumatoid arthritis, asthma, dermatitis, and psoriasis. These trials, however, documented only moderate beneficial effects of H4R ligands so far. Nevertheless, pre-clinically, H4R still is subject of ongoing research, analyzing various inflammatory, allergic, and autoimmune diseases. During inflammatory reactions in gut tissues, histamine concentrations rise in affected areas, indicating its possible biological effect. Indeed, in histamine-deficient mice experimentally induced inflammation of the gut is reduced in comparison to that in histamine-competent mice. However, antagonists at H1R, H2R, and H3R do not provide an effect on inflammation, supporting the idea that H4R is responsible for the histamine effects. In the present review, we discuss the involvement of histamine and H4R in inflammatory and inflammation-associated diseases of the gut.
Collapse
|
19
|
Lin W, Li Q, Zhang D, Zhang X, Qi X, Wang Q, Chen Y, Liu C, Li H, Zhang S, Wang Y, Shao B, Zhang L, Yuan Q. Mapping the immune microenvironment for mandibular alveolar bone homeostasis at single-cell resolution. Bone Res 2021; 9:17. [PMID: 33723232 PMCID: PMC7960742 DOI: 10.1038/s41413-021-00141-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/18/2021] [Accepted: 02/01/2021] [Indexed: 02/04/2023] Open
Abstract
Alveolar bone is the thickened ridge of jaw bone that supports teeth. It is subject to constant occlusal force and pathogens invasion, and is therefore under active bone remodeling and immunomodulation. Alveolar bone holds a distinct niche from long bone considering their different developmental origin and postnatal remodeling pattern. However, a systematic explanation of alveolar bone at single-cell level is still lacking. Here, we construct a single-cell atlas of mouse mandibular alveolar bone through single-cell RNA sequencing (scRNA-seq). A more active immune microenvironment is identified in alveolar bone, with a higher proportion of mature immune cells than in long bone. Among all immune cell populations, the monocyte/macrophage subpopulation most actively interacts with mesenchymal stem cells (MSCs) subpopulation. Alveolar bone monocytes/macrophages express a higher level of Oncostatin M (Osm) compared to long bone, which promotes osteogenic differentiation and inhibits adipogenic differentiation of MSCs. In summary, our study reveals a unique immune microenvironment of alveolar bone, which may provide a more precise immune-modulatory target for therapeutic treatment of oral diseases.
Collapse
Affiliation(s)
- Weimin Lin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiwen Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Danting Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaohan Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingying Qi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qian Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yaqian Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Caojie Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hanwen Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuan Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Shao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China. .,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
20
|
Gruenbacher G, Gander H, Dobler G, Rahm A, Klaver D, Thurnher M. The human G protein-coupled ATP receptor P2Y 11 is a target for anti-inflammatory strategies. Br J Pharmacol 2021; 178:1541-1555. [PMID: 33463722 PMCID: PMC9328440 DOI: 10.1111/bph.15379] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 12/24/2022] Open
Abstract
Background and Purpose The ATP receptor P2Y11, which couples to Gq and Gs proteins, senses cell stress and promotes cytoprotective responses. P2Y11 receptors are upregulated during differentiation of M2 macrophages. However, it is unclear whether and how P2Y11 receptors contribute to the anti‐inflammatory properties of M2 macrophages. Experimental Approach Transcriptome and secretome profiling of ectopic P2Y11 receptors was used to analyse their signalling and function. Findings were validated in human monocyte‐derived M2 macrophages. The suramin analogue NF340 and P2Y11 receptor‐knockout cells confirmed that agonist‐mediated responses were specific to P2Y11 receptor stimulation. Key Results Temporal transcriptome profiling of P2Y11 receptor stimulation showed a strong and tightly controlled response of IL‐1 receptors, including activation of the IL‐1 receptor target genes, IL6 and IL8. Secretome profiling confirmed the presence of IL‐6 and IL‐8 proteins and additionally identified soluble tumour necrosis factor receptor 1 and 2 (sTNFR1 and sTNFR2) as targets of P2Y11 receptor activation. Raised levels of intracellular cAMP in M2 macrophages, after inhibition of phosphodiesterases (PDE), especially PDE4, strongly increased P2Y11 receptor‐induced release of sTNFR2 through ectodomain shedding mediated by TNF‐α converting enzyme (TACE/ADAM17). Both IL‐1α and IL‐1ß synergistically enhanced P2Y11 receptor‐ induced IL‐6 and IL‐8 secretion and release of sTNFR2. During lipopolysaccharide‐induced activation of TLR4, which shares the downstream signalling pathway with IL‐1 receptors, P2Y11 receptors specifically prevented secretion of TNF‐α. Conclusions and Implications Targeting P2Y11 receptors activates IL‐1 receptor signalling to promote sTNFR2 release and suppress TLR4 signalling to prevent TNF‐α secretion, thus facilitating resolution of inflammation.
Collapse
Affiliation(s)
- Georg Gruenbacher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hubert Gander
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Dobler
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Rahm
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Klaver
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
21
|
Li Y, Feng J, Song S, Li H, Yang H, Zhou B, Li Y, Yue Z, Lian H, Liu L, Hu S, Nie Y. gp130 Controls Cardiomyocyte Proliferation and Heart Regeneration. Circulation 2020; 142:967-982. [PMID: 32600062 DOI: 10.1161/circulationaha.119.044484] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background:
A key cause of the high mortality of cardiovascular diseases is the cardiomyocyte inability to renew after cardiac injury. As a promising strategy to supplement functional myocytes for cardiac repair, there is a pressing need to understand the cellular and molecular mechanisms of heart regeneration.
Methods:
Seven genetic mouse lines were used: global OSM (oncostatin M) knockout, monocyte-/macrophage-specific OSM deletion, cardiomyocyte-specific lines, including OSM receptor deletion, gp130 (glycoprotein 130) deletion, gp130 activation, and Yap (yes-associated protein) ablation with gp130 activation mice. A series of molecular signaling experiments, including RNA sequencing, immunostaining, coimmunoprecipitation, and imaging flow cytometry, were conducted. Two models of cardiac injury, apical resection and myocardial infarction operation, were performed in neonatal, juvenile, and adult mice. Heart regeneration and cardiac function were evaluated by Masson staining and echocardiography, respectively. Gene recombinant adenovirus-associated virus was constructed and infected myocardial-infarcted mice as a gene therapy.
Results:
OSM was identified by RNA sequencing as a key upstream regulator of cardiomyocyte proliferation during neonatal heart regeneration in mice. Cardiomyocyte proliferation and heart regeneration were suspended in neonatal mice after cardiac injury when OSM was conditionally knockout in macrophages. The cardiomyocyte-specific deficiency of the OSM receptor heterodimers, OSM receptor and gp130, individually in cardiomyocytes reduced myocyte proliferation and neonatal heart regeneration. Conditional activation of gp130 in cardiomyocytes promoted cardiomyocyte proliferation and heart regeneration in juvenile and adult mice. Using RNA sequencing and functional screening, we found that Src mediated gp130-triggered cardiomyocyte proliferation by activating Yap (yes-associated protein) with Y357 phosphorylation independently of the Hippo pathway. Cardiomyocyte-specific deletion of Yap in
Myh6-gp130
ACT
mice blocked the effect of gp130 activation–induced heart regeneration in juvenile mice. Gene therapy with adenovirus-associated virus encoding constitutively activated gp130 promoted cardiomyocyte proliferation and heart regeneration in adult mice after myocardial infarction.
Conclusions:
Macrophage recruitment is essential for heart regeneration through the secretion of OSM, which promotes cardiomyocyte proliferation. As the coreceptor of OSM, gp130 activation is sufficient to promote cardiomyocyte proliferation by activating Yap through Src during heart regeneration. gp130 is a potential therapeutic target to improve heart regeneration after cardiac injury.
Collapse
Affiliation(s)
- Yandong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Jie Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Haotong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Huijun Yang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (H.Y.)
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Shanghai (B.Z.)
| | - Yan Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China (Yan Li)
| | - Zhang Yue
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.Y.)
| | - Hong Lian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Lihui Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| |
Collapse
|
22
|
Chazot PL, Tiligada E. Challenges in the development and exploitation of new therapeutic options targeting the histaminergic system. Br J Pharmacol 2020; 177:467-468. [PMID: 31994717 PMCID: PMC7012940 DOI: 10.1111/bph.14947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
LINKED ARTICLES This article is part of a themed section on New Uses for 21st Century Antihistamines. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.3/issuetoc.
Collapse
Affiliation(s)
| | - Ekaterini Tiligada
- Department of Pharmacology, Medical SchoolNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
23
|
Mommert S, Hüer M, Schaper-Gerhardt K, Gutzmer R, Werfel T. Histamine up-regulates oncostatin M expression in human M1 macrophages. Br J Pharmacol 2019; 177:600-613. [PMID: 31328788 PMCID: PMC7012943 DOI: 10.1111/bph.14796] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 05/14/2019] [Accepted: 07/07/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Human monocyte-derived M1 macrophages develop in relation to growth factors, bacterial products, and cytokines in a local micro-environment. M1 macrophages produce pro-inflammatory mediators, in particular, oncostatin M (OSM), which is secreted from the cells in response to the active complement component C5a. As C5a also releases histamine from human mast cells and shows immune modulatory functions similar to histamine in regulating expression of the IL-12 cytokine family, we investigated the effects of histamine on OSM expression in human M1 macrophages. EXPERIMENTAL APPROACH Cytokine expression was analysed by real-time quantitative PCR and elisa techniques. Normal human epidermal keratinocytes were stimulated with supernatants from activated M1 macrophages, and phosphorylation of STAT3 was assessed by flow cytometry. KEY RESULTS OSM mRNA expression was highly up-regulated by histamine and agonists targeting the histamine H1 H2 , and H4 receptors in human M1 macrophages and by C5a, which was used as control stimulus. Protein levels of OSM and IL-6 were up-regulated by histamine. Supernatants from histamine-stimulated, fully differentiated M1 macrophages were able to phosphorylate STAT3 in normal human epidermal keratinocytes. CONCLUSIONS AND IMPLICATIONS The up-regulation of OSM expression in response to histamine and C5a shown in this study provides further evidence that histamine and C5a, acting through their GPCRs, have almost equal functional effects in cells of the monocyte lineage. Both mediators OSM and IL-6 have the capability to activate human keratinocytes. This effect may have an influence on the course of inflammatory skin diseases. LINKED ARTICLES This article is part of a themed section on New Uses for 21st Century. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.3/issuetoc.
Collapse
Affiliation(s)
- Susanne Mommert
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Marius Hüer
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Katrin Schaper-Gerhardt
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Ralf Gutzmer
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| |
Collapse
|