1
|
Xu Z, Wang Y, Li S, Li Y, Chang L, Yao Y, Peng Q. Advances of functional nanomaterials as either therapeutic agents or delivery systems in the treatment of periodontitis. BIOMATERIALS ADVANCES 2025; 175:214326. [PMID: 40300444 DOI: 10.1016/j.bioadv.2025.214326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Periodontitis is a common chronic inflammatory disease primarily caused by pathogenic microorganisms in the oral cavity. Without appropriate treatments, it may lead to the gradual destruction of the supporting tissues of the teeth. While current treatments can alleviate symptoms, they still have limitations, particularly in eliminating pathogenic bacteria, promoting periodontal tissue regeneration, and avoiding antibiotic resistance. In recent years, functional nanomaterials have shown great potential in the treatment of periodontitis due to their unique physicochemical and biological properties. This review summarizes various functionalization strategies of nanomaterials and explores their potential applications in periodontitis treatment, including metal-based nanoparticles, carbon nanomaterials, polymeric nanoparticles, and exosomes. The mechanisms and advances in antibacterial effects, immune regulation, reactive oxygen species (ROS) scavenging, and bone tissue regeneration are discussed in detail. In addition, the challenges and future directions of applying nanomaterials in periodontitis therapy are also discussed.
Collapse
Affiliation(s)
- Ziyi Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yue Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shuoshun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuanhong Li
- Department of Orthodontics, Shanghai Stomatological Hospital and School of Stomatology, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, China
| | - Lili Chang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Long L, Li M, Wang M, Liang B, Huang M, Yuan X, Wu X, Guo X, Li S, Liu Z, Liu W, Chen W, Wang W, Lyu Q, Li C. Activation of mannose receptor C type 1 in macrophages improves renal fibrosis through mediating fibronectin endocytosis. Life Sci 2025; 371:123593. [PMID: 40164332 DOI: 10.1016/j.lfs.2025.123593] [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: 01/21/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
AIMS Excess extracellular matrix (ECM) deposition is the characteristic of renal fibrosis, owing to the imbalance between synthesis and degradation. Fibronectin could regulate the deposition of other ECM, thus plays a crucial role in the progression of renal fibrosis. Mannose receptor C type 1 (MRC1), largely expressed on macrophages, owns an extracellular fibronectin type II domain that binds to and internalizes collagen and thus involves in fibrosis modulation. The purpose of the present study was to investigate whether MRC1 participates in the internalization of fibronectin and whether alginate oligosaccharides (AOSC), a degradation product of alginate, has beneficial effects in the resolution of renal fibrosis via MRC1. MATERIALS AND METHODS Renal fibrosis models were constructed by unilateral ureteral obstruction (UUO) and unilateral ischemia-reperfusion injury (UIRI) in MRC1-WT and MRC1-KO mice. RAW264.7 cells were treated with TGF-β1 to induce pro-fibrotic responses. Expression of fibrotic markers and fibronectin endocytosis were examined. KEY FINDINGS MRC1 gene knockout aggravated renal fibrosis in UUO and UIRI models. Inhibition of MRC1 exacerbated TGF-β1-induced pro-fibrotic responses in RAW264.7 cells. MRC1 regulated integrin β1-mediated fibronectin endocytosis through Arp2/3-Kindlin-2 signaling pathway. AOSC improved renal fibrosis by increasing MRC1 expression and endocytosis of fibronectin. SIGNIFICANCE Our findings highlight the importance of MRC1 and fibronectin endocytosis in the development of renal fibrosis, suggesting that activation of MRC1 by AOSC is probably a therapeutic option to delay the progress of kidney fibrosis.
Collapse
Affiliation(s)
- Luosha Long
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathology and Pathophysiology, Pu Ai Medical School, Shaoyang University, Shaoyang, China
| | - Meng Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Minghui Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Baien Liang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Meiying Huang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xi Yuan
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinyan Wu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiangdong Guo
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Suchun Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, China
| | - Zhende Liu
- Haitang (Jiangsu) Biotechnology Co, Ltd., Nantong, Jiangsu, China
| | - Weizhi Liu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qianqian Lyu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| | - Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Liu Y, Terracciano R, Scheerstra J, Yilmaz G, Wu H, Welzen P, Cao S, Padial TP, Abdelmohsen L, Shao J, Sun B, Becer CR, van Hest JCM. Fast and Autonomous Mannosylated Nanomotors for Dynamic Cancer Cell Targeting. Angew Chem Int Ed Engl 2025; 64:e202505717. [PMID: 40169377 PMCID: PMC12124440 DOI: 10.1002/anie.202505717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/03/2025]
Abstract
An attractive strategy in cancer cell therapy is to employ motile nanoparticles that can actively search for their target. Herein, we introduce mannosylated compartmentalized cross-linked enzyme-driven nanomotors (c-CLEnM), which exhibit specific and efficient targeting of Hep G2 cells through elevated autonomous motion. In this design, we constructed biodegradable bowl-shaped stomatocytes encapsulating the enzymes glucose oxidase (GOx) and catalase (CAT) within their nanocavity. A subsequent enzyme crosslinking reaction was performed to guarantee their stability. Furthermore, the c-CLEnM were surface modified with a mannose-functional glycopolymer, enabling binding with receptors expressed on Hep G2 cells. Interestingly, the targeting ligands on the nanomotors not only improved their specificity toward cancer cells but also enhanced motility. Compared to the non-mannosylated nanomotors, mannosylated c-CLEnM exhibited enhanced motion and higher targeting efficiency to cells in glucose-containing ionic environments. The unexpected acceleration in speed resulted from the surface modification of these nanomotors with a glycopolymer layer, which increased the zeta potential and created a shielding effect that mitigated the influence of the surrounding ions. This nanomotor design highlights the synergistic effect of functional glycopolymer modification on cellular uptake, adding an additional level of control to nanomotors for application in cancer therapy.
Collapse
Affiliation(s)
- Yuechi Liu
- Bio‐Organic ChemistryInstitute of Complex Molecular SystemsEindhoven University of TechnologyHelix, P. O. Box 513Eindhoven5600 MBthe Netherlands
| | | | - Jari Scheerstra
- Bio‐Organic ChemistryInstitute of Complex Molecular SystemsEindhoven University of TechnologyHelix, P. O. Box 513Eindhoven5600 MBthe Netherlands
| | - Gokhan Yilmaz
- Department of ChemistryUniversity of WarwickCoventryCV4 7ALUK
| | - Hanglong Wu
- Bio‐Organic ChemistryInstitute of Complex Molecular SystemsEindhoven University of TechnologyHelix, P. O. Box 513Eindhoven5600 MBthe Netherlands
| | - Pascal Welzen
- Bio‐Organic ChemistryInstitute of Complex Molecular SystemsEindhoven University of TechnologyHelix, P. O. Box 513Eindhoven5600 MBthe Netherlands
| | - Shoupeng Cao
- College of Polymer Science and EngineeringSichuan UniversityChengdu610065P.R. China
| | - Tania Patino Padial
- Bio‐Organic ChemistryInstitute of Complex Molecular SystemsEindhoven University of TechnologyHelix, P. O. Box 513Eindhoven5600 MBthe Netherlands
| | - Loai Abdelmohsen
- Bio‐Organic ChemistryInstitute of Complex Molecular SystemsEindhoven University of TechnologyHelix, P. O. Box 513Eindhoven5600 MBthe Netherlands
| | - Jingxin Shao
- Bio‐Organic ChemistryInstitute of Complex Molecular SystemsEindhoven University of TechnologyHelix, P. O. Box 513Eindhoven5600 MBthe Netherlands
| | - Bingbing Sun
- Bio‐Organic ChemistryInstitute of Complex Molecular SystemsEindhoven University of TechnologyHelix, P. O. Box 513Eindhoven5600 MBthe Netherlands
| | - C. Remzi Becer
- Department of ChemistryUniversity of WarwickCoventryCV4 7ALUK
| | - Jan C. M. van Hest
- Bio‐Organic ChemistryInstitute of Complex Molecular SystemsEindhoven University of TechnologyHelix, P. O. Box 513Eindhoven5600 MBthe Netherlands
| |
Collapse
|
4
|
Jain M, Jadhav IM, Dangat SV, Singuru SR, Sethi G, Yuba E, Gupta RK. Overcoming the novel glycan-lectin checkpoints in tumor microenvironments for the success of the cross-presentation-based immunotherapy. Biomater Sci 2025. [PMID: 40421610 DOI: 10.1039/d4bm01732c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
In pursuit of meeting the ever-rising demand for cancer therapies, cross-presentation-based glyconanovaccines (GNVs) targeting C-type lectin receptors (CLRs) on DCs have shown significant potential as cutting-edge cancer immunotherapy. GNVs are an attractive approach to induce anti-cancer cytotoxic T lymphocyte responses. Despite immune checkpoints (ICs) being well established and an obstacle to the success of GNVs, glycan-lectin circuits are emerging as unique checkpoints due to their immunomodulatory functions. Given the role of aberrant tumor glycosylation in promoting immune evasion, mitigating these effects is crucial for the efficacy of GNVs. Lectins, such as siglecs and galectins, are detrimental to the tumor immune landscape as they promote an immunosuppressive TME. From this perspective, this review aims to explore glycan-lectin ICs and their influence on the efficacy of GNVs. We aim to discuss various ICs in the TME followed by drawbacks of immune checkpoint inhibitors (ICIs). We will also emphasize the altered glycosylation profile of tumors, addressing their immunosuppressive nature along with ways in which CLRs, siglecs, and galectins contribute to immune evasion and cancer progression. Considering the resistance towards ICIs, current and prospective approaches for targeting glycan-lectin circuits and future prospects of these endeavors in harnessing the full potential of GNVs will also be highlighted.
Collapse
Affiliation(s)
- Mannat Jain
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Isha M Jadhav
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Suyash Vinayak Dangat
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Srinivasa Rao Singuru
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Eiji Yuba
- Department of Chemistry & Bioengineering, Graduate School of Engineering, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka-city, Osaka 558-8585, Japan.
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| |
Collapse
|
5
|
Moosavi M, Brødsgaard Kjærup R, Papanikolaou K, Wattrang E, Sørensen Dalgaard T. Indications of trained innate immunity by Escherichia coli vaccination or chitin feed supplementation assessed during Ascaridia galli infection in chickens. Mol Immunol 2025; 183:246-258. [PMID: 40414091 DOI: 10.1016/j.molimm.2025.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 05/04/2025] [Accepted: 05/10/2025] [Indexed: 05/27/2025]
Abstract
Infections with the gastrointestinal roundworm Ascaridia galli, cause health problems and economic losses in laying hen husbandry, particularly in organic and free-range systems. This study aimed to evaluate induction of trained innate immunity through priming with a live attenuated Escherichia coli vaccine or chitin supplementation in the feed as a novel approach to mitigate A. galli infection. The study comprised four groups of chickens: chitin-fed (day 1-7 of age), E. coli-vaccinated (day 1 of age), an untreated control group, and a naïve uninfected group. On day 7 of age, the first three groups were infected with A. galli. Immune parameters were assessed after initial treatments and post the parasite infection. Also, faecal excretion of nematode eggs and total worm burden were monitored post-infection. The chitin and E. coli treatments induced changed proportions of leukocytes in bone marrow as well as changes in cell surface receptor expression. Moreover, treatments altered the immune response to the A. galli infection, e.g. observed for numbers of heterophils and TCRγδ+CD8- T-cells in the circulation but also expression levels of cell surface receptors CD41/61, Bu-1 and MHC-II on circulating leukocyte subsets. However, neither treatment affected worm burden, faecal egg excretion or the induction of A. galli-specific IgY. The results demonstrate potential in vivo training of the avian innate immune system but further research is needed to identify strategies to explore this in relation to control of nematode infections.
Collapse
Affiliation(s)
- Mona Moosavi
- Department of Animal and Veterinary Sciences, Aarhus University, Tjele, Denmark; Department of Poultry Science at Tarbiat Modares University, Tehran, Iran
| | | | | | - Eva Wattrang
- Department of Microbiology, Swedish Veterinary Agency, Uppsala, Sweden
| | | |
Collapse
|
6
|
Yang G, Tan W, Yan L, Lao Q, Zheng W, Ding H, Yu J, Liu Y, Zou L, Guo M, Yu L, Zhou X, Li W, Yang L. Phillyrin for sepsis-related acute lung injury: A potential strategy suppressing GSK-3β. Mol Immunol 2025; 183:115-136. [PMID: 40359720 DOI: 10.1016/j.molimm.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
The efficacy of clinical drugs for acute lung injury/acute respiratory distress syndrome (ALI/ARDS) remains suboptimal. Phillyrin (PHN), a compound derived from Forsythia, is believed to alleviate sepsis-related ALI/ARDS; however, its mechanisms are not fully elucidated. In this study, we screened 8331 target genes associated with ALI/ARDS from public databases and identified six hub genes relevant to PHN treatment: AKT1, GSK-3β, PPP2CA, PPP2CB, PPP2R1A, and AR. Receiver operating characteristic analysis and single-cell sequencing analysis revealed the expression of AKT1, GSK-3β, PPP2CA, PPP2CB, and PPP2R1A were markedly elevated. Molecular docking and dynamics simulations indicated that PHN forms a structurally stable complex with glycogen synthase kinase-3β (GSK-3β). Mendelian randomization analyses suggested that PHN, as a potent GSK-3β inhibitor, may promote M2 macrophage polarization and reduce neutrophil recruitment. We validated these findings through in vivo and in vitro experiments, demonstrating that PHN lowers iNOS levels and raises MMR levels by downregulating GSK-3β mRNA expression and protein activity during lipopolysaccharide (LPS)-induced macrophage inflammation. Additionally, PHN inhibited GSK-3β mRNA expression and protein activity, reducing NF-κB-p65 nuclear translocation in LPS-induced zebrafish inflammation and mice ALI. This inhibition decreased levels of TNF-α and IL-6, increased IL-10 levels, promoted M2 macrophage polarization, suppressed neutrophil recruitment, and ultimately ameliorated ALI/ARDS. In conclusion, our results indicate that PHN effectively alleviates LPS-induced ALI/ARDS by suppressing GSK-3β signaling.
Collapse
Affiliation(s)
- Guangli Yang
- Department of Central Laboratory, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Weifu Tan
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Lijun Yan
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qiaocong Lao
- Central Laboratory, The Tenth Affiliated Hospital, Southern Medical University, Dongguan People's Hospital, Dongguan 523059, China
| | - Wujuan Zheng
- Department of Pharmacy, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Hongyan Ding
- Omega-3 Research and Conversion Center, Dongguan Innovation Research Institute, Guangdong Medical University, Dongguan 523900, China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yong Liu
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Liyi Zou
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Maorun Guo
- Pingyi Health Center of Pingyi County, Linyi 273300, China
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiangjun Zhou
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Wei Li
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China.
| | - Liling Yang
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China; Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Pharmacy, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China.
| |
Collapse
|
7
|
Liu T, Gu X, Qin Z, Zhang S, Yang R, Huang Y, Yan Y, Zhu P, Seeberger PH, Zhu Y. Novel Base-Labile Linkers for Convenient Purification and Functional Group Introduction in Solid-Phase Glycan Synthesis. Chemistry 2025; 31:e202500603. [PMID: 40035230 DOI: 10.1002/chem.202500603] [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: 02/16/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/05/2025]
Abstract
Glycans are essential biomolecules across species, but their structure-property relationships remain poorly understood. Pure oligosaccharide samples are crucial for studying these properties, and their preparation heavily relies on chemical synthesis. The linker used for attaching the oligosaccharide to the solid support is essential during solid-phase glycan synthesis. Here, we present three base-labile linkers compatible with Merrifield resin. The first type of resin, containing a hydroxyhexanoic acid linker, can be prepared in a one-step synthesis and releases glycans with a terminal carboxylic acid after methanolysis and hydrolysis. Linkers on the second and third types of resin can be synthesized through simple chemical reactions and cleaved to reveal either a free reducing end of the oligosaccharide or a terminal amine with a C5-spacer. The phthalate structure in the second and third types of resin is sensitive to sodium methoxide and can also be cleaved during hydrazinolysis. Hydrazinolysis, rather than methanolysis, enables the isolation of oligosaccharide with pivaloyl ester protection and facilitates the purification process compared to partially protected compounds. Using these resins, we prepared nine tetrasaccharides and one octasaccharide, including α (1-6) mannan, α (1-4) glucan, β (1-4) glucan, and β (1-3) glucan. Glycans with terminal functional groups can be converted into glycoconjugates for further applications.
Collapse
Affiliation(s)
- Tan Liu
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Xin Gu
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
- Changzhou Hospital of Traditional Chinese Medicine, Changzhou, 213003, China
| | - Zimiao Qin
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Size Zhang
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Runxin Yang
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Yi Huang
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Yishu Yan
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Pan Zhu
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Peter H Seeberger
- Department of Biomolecular Systems, Max-Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476, Potsdam, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany
| | - Yuntao Zhu
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
8
|
Zhu XR, Tang WH, Peng Q, Zhang R, Chen YH, Liu QN, Tang BP, Dai LS. Comprehensive analysis of the transcriptome implicated in the immune response of Procambarus clarkii to Aeromonas hydrophila. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 56:101528. [PMID: 40408924 DOI: 10.1016/j.cbd.2025.101528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/16/2025] [Accepted: 05/05/2025] [Indexed: 05/25/2025]
Abstract
In order to better understand the immune response of Procambarus clarkii to Aeromonas hydrophila injection, the transcriptome data of the gill tissue of P. clarkii were compared and analyzed. 1008 significant DEGs were identified in A. hydrophila infected and PBS control groups, including 411 up-regulated genes and 597 down-regulated genes. Endocytosis, phagocyte and lysosome were the most clustered pathways of DEGs in KEGG database. RNA-Seq results were validated through the verification of immune-related differentially expressed genes (DEGs), whose expression levels were assessed using quantitative real-time PCR (qRT-PCR). With the increase of treatment time of A. hydrophila, the total protein in gill of the treatment group showed a trend of increasing first, then decreasing and then increasing and decreasing. ACP and AKP both show a trend of rising first, then falling and then rising. The results of comprehensive research showed that crayfish infected with A. hydrophila caused damage to gill tissue, and the related immune genes were up-regulated and the immune mechanism was operated to protect the body from A. hydrophila. The differential gene MRC1 was screened through the transcriptome, and to further understand its impact, it was disrupted by RNAi technology, which showed a significant down-regulation of immune genes (TAB2, TLR3, ALF6, Lyso3, clotting factor G beta subunit-like and coagulation factor X-like) as well as genes downstream of the pathway (AP4E1, ARSB and TUBA1A). This study provides a theoretical basis for further exploring the immune adaptability of aquatic animals under bacterial infection.
Collapse
Affiliation(s)
- Xi-Rong Zhu
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China; College of Biotechnology and Pharmaceutical Engineering, Nanjing University of Technology, Nanjing 210009, People's Republic of China
| | - Wen-Hui Tang
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China
| | - Qin Peng
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China
| | - Rui Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, People's Republic of China
| | - Yao-Hui Chen
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China
| | - Qiu-Ning Liu
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China.
| | - Bo-Ping Tang
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China.
| | - Li-Shang Dai
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, People's Republic of China
| |
Collapse
|
9
|
Liu K, Tian X, Fei S, Song Y, Abd El-Aty AM, Tan M. Macrophage and mitochondrion dual-targeting astaxanthin nanoparticles prepared by Maillard reaction for colonic inflammation alleviation. MARINE LIFE SCIENCE & TECHNOLOGY 2025; 7:352-365. [PMID: 40417255 PMCID: PMC12102426 DOI: 10.1007/s42995-024-00255-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 08/21/2024] [Indexed: 05/27/2025]
Abstract
This study demonstrated the design of whey protein isolate (WPI)-mannose (Man) conjugates with triphenylphosphonium bromide (TPP) through self-assembly to prepare macrophage and mitochondrion dual-targeting astaxanthin (AXT) nanoparticles (AXT@TPP-WPI-Man). The nanoparticles displayed spherical structures with a well-dispersed size of approximately 206.1 ± 39.2 nm, with good biocompatibility, stability, and targeting capabilities. In vitro experiments demonstrated the specific accumulation of AXT@TPP-WPI-Man in mitochondria and exhibited good targeting ability toward macrophages. The AXT@TPP-WPI-Man effectively reduced reactive oxygen species and preserved the normal mitochondrial membrane potential. The AXT@TPP-WPI-Man treated ulcerative colitis mice exhibited a 52.32% increase in colon length with significant improvement in weight loss, disease activity index scores, and reduced release of inflammatory cytokines. Immunofluorescence staining indicated AXT@TPP-WPI-Man alleviated ulcerative colitis by reducing M1 polarization in colonic macrophages while promoting M2 polarization. The dual-targeting AXT@TPP-WPI-Man has the potential to improve astaxanthin bioavailability, presenting a promising delivery method for the treatment of ulcerative colitis. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-024-00255-9.
Collapse
Affiliation(s)
- Kangjing Liu
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| | - Xueying Tian
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| | - Siyuan Fei
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| | - Yukun Song
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| | - A. M. Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211 Egypt
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, 25240 Erzurum, Turkey
| | - Mingqian Tan
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| |
Collapse
|
10
|
Swami R, Popli P, Sal K, Challa RR, Vallamkonda B, Garg M, Dora CP. A review on biomacromolecular ligand-directed nanoparticles: New era in macrophage targeting. Int J Biol Macromol 2025; 306:141740. [PMID: 40058437 DOI: 10.1016/j.ijbiomac.2025.141740] [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: 06/06/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 05/11/2025]
Abstract
Traditional drug delivery strategies often have side effects due to uneven drug distribution leading to the subtherapeutic impacts. Ligand-modified nanoparticles offer a revolutionary approach to precise drug delivery. These modified nanoparticles can potentially target macrophages, which is crucial for defense and disease progression efficiently. Out of many classes of ligands, biomacromolecular ligands emerged as potential ligands for directing these nanoparticles to macrophages due to their consecutive receptors over the macrophage surface, assisting easy internalization and thus supporting elevated efficacy and reduced toxicity. This approach could significantly improve treatment for diseases like cancer, tuberculosis, etc. by directing drugs to macrophages and reducing side effects. By leveraging nanotechnology and biomacromolecular-based ligand-directed targeting, we can achieve more precise and effective treatments, paving the way for advancements in precision medicine.
Collapse
Affiliation(s)
- Rajan Swami
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Pankaj Popli
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Komal Sal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | | | - Madhukar Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
11
|
Kou H, Qi L, Huang D, Wu J, Shi H, Yang H. Scalable Fabrication of High-Payload Dendrimer-Based Nanoparticles for Targeted Atherosclerosis Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:24953-24962. [PMID: 40237537 DOI: 10.1021/acsami.5c00816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Nanoparticle-based therapeutics hold promise for the treatment of atherosclerosis, but challenges such as low drug-loading capacity and a lack of scalable, controllable production hinder their clinical translation. Flash nanoprecipitation, a continuous synthesis method, offers a potential solution for scalable and reproducible nanoparticle production. In this study, we employed a custom-designed multi-inlet vortex mixer to perform cross-linking reaction-enabled flash nanoprecipitation, facilitating controlled and scalable synthesis of cross-linked polyamidoamine (PAMAM) dendrimer nanoparticles. Notably, this approach allows simultaneous nanoparticle cross-linking and drug loading in a single step. The mannose moiety enabled specific targeting of macrophages via mannose receptors, enhancing the localization of the nanoparticles to atherosclerotic plaques. Atorvastatin calcium, a widely used clinical drug for atherosclerosis treatment, was selected as the model drug. This approach achieved both high production rates and high drug-loading capacities, with an output flow rate of 9.6 L/h and a nanoparticle concentration of approximately 0.4 g/L. The optimized formulation exhibited a drug-loading capacity of 37% and an encapsulation efficiency of 76%. In vitro and in vivo experiments demonstrated effective macrophage and plaque targeting, leading to significant therapeutic benefits. Treatment with these nanoparticles resulted in approximately 40% inhibition of aortic root plaque progression compared to the free drug-treated group. This scalable and efficient nanoparticle platform is a promising strategy for improving atherosclerosis treatment.
Collapse
Affiliation(s)
- Huari Kou
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Lin Qi
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Da Huang
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jiandong Wu
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Honglan Shi
- Department of Chemistry, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Hu Yang
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| |
Collapse
|
12
|
Liang BE, Long LS, Wu XY, Huang MY, Lai Y, Yuan X, Wang MH, Li M, Zheng QQ, Zhang HL, Chen MC, Liu ZD, Geng X, Lyu QQ, Wang WD, Liu QH, Liu WZ, Li CL. Alginate oligosaccharide prevents renal ischemia-reperfusion injury in rats via MRC1-mediated pathway. Acta Pharmacol Sin 2025:10.1038/s41401-025-01545-3. [PMID: 40263568 DOI: 10.1038/s41401-025-01545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 04/24/2025]
Abstract
Acute kidney injury (AKI) is a clinical syndrome that is defined as a sudden decline in renal function and characterized by inflammation and tubular injury. Alginate oligosaccharide (AOSC), a natural product obtained from alginate by acidolysis and hydrolysis, shows activities of antioxidant, immunomodulation, and anti-inflammation. In this study, we investigated the potential of AOSC in the treatment of AKI. Renal ischemia-reperfusion (I/R) was induced in male rats by clipping both the renal artery and vein for 45 min followed by reperfusion for 24 h. The rats were treated with AOSC (100 mg/kg, i.g.) before surgery. At the end of the experiments, both kidneys were collected for protein, mRNA measurement, or histological analysis. We showed that AOSC pretreatment significantly improved glomerular and tubular function in the kidney of I/R rats. AOSC markedly inhibited I/R-induced activation of TLR4/MyD88/NF-κB/IL-1β inflammatory signaling and prevented apoptosis in the kidney. In HK2 cells subjected to hypoxia/reoxygenation (H/R) stimulation, AOSC (250-1000 μg/ml) dose-dependently prevented pro-inflammatory responses and cell apoptosis. Transcriptomic analysis revealed that I/R increased the expression levels of mannose receptor type C1 (MRC1) in the kidney, which was markedly inhibited by AOSC. Molecular docking showed that AOSC interacted with E725, N727, E733, T743, S745, and N747 of MRC1 through hydrogen bonds. MRC1 gene knockout significantly improved renal function and attenuated I/R-induced kidney inflammation and apoptosis in mice. In line with this, AOSC failed to prevent I/R-induced kidney injury in MRC1 gene knockout mice. UPLC analysis showed that the protection of AOSC in HK2 cells subjected to H/R was likely attributed to MRC1-mediated intracellular endocytosis. In conclusion, AOSC prevents I/R-induced AKI, which is at least partially mediated by MRC1.
Collapse
Affiliation(s)
- Bai-En Liang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Luo-Sha Long
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin-Yan Wu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mei-Ying Huang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying Lai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xi Yuan
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ming-Hui Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Meng Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qi-Qi Zheng
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hai-Ling Zhang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Man-Chun Chen
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhen-de Liu
- Haitang (Jiangsu) Biotechnology Co Ltd, Nantong, 226100, China
| | - Xin Geng
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Qian-Qian Lyu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Wei-Dong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing-Hua Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
- Department of Nephrology, Jieyang People's Hospital, Jieyang, 522000, China.
| | - Wei-Zhi Liu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Chun-Ling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
13
|
Jonsson A, Korsgren O, Hedin A. Transcriptomic characterization of human pancreatic CD206- and CD206 + macrophages. Sci Rep 2025; 15:12037. [PMID: 40199933 PMCID: PMC11978877 DOI: 10.1038/s41598-025-96313-y] [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: 11/26/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Macrophages reside in all organs and participate in homeostatic- and immune regulative processes. Little is known about pancreatic macrophage gene expression. In the present study, global gene expression was characterized in human pancreatic macrophage subpopulations. CD206- and CD206 + macrophages were sorted separately from pancreatic islets and exocrine tissue to high purity using flow cytometry, followed by RNA-seq analysis. Comparing CD206- with CD206 + macrophages, CD206- showed enrichment in histones, proliferation and cell cycle regulation, glycolysis and SPP1-associated immunosuppressive polarization while CD206 + showed enrichment in complement and coagulation-, IL-10 and IL-2RA immune regulation, as well as scavenging-related gene sets. Comparing islet CD206- with exocrine CD206-, enrichments in islet samples included two sets involved in immune regulation, while enrichments in exocrine samples included sets related to extracellular matrix and immune activation. Fewer differences were found between CD206 + macrophages, with enrichments in islet samples including two IL2-RA related gene sets, while enrichments in exocrine samples included sets related to extracellular matrix and immune activation. Comparing macrophages between individuals with normoglycemia, elevated HbA1c or type 2 diabetes, only a few diverse differentially expressed genes were identified. This work characterizes global gene expression and identifies differences between CD206- and CD206 + macrophage populations within the human pancreas.
Collapse
Affiliation(s)
- Alexander Jonsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Anders Hedin
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
14
|
Sarsarshahi S, Bhattacharya S, Zacharias ZR, Kamel ES, Houtman JCD, Nejadnik R. Highly variable aggregation and glycosylation profiles and their roles in immunogenicity to protein-based therapeutics. J Pharm Sci 2025; 114:103771. [PMID: 40139530 DOI: 10.1016/j.xphs.2025.103771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Production of antibodies against protein-based therapeutics (e.g., monoclonal antibodies (mAbs)) by a recipient's immune system can vary from benign symptoms to chronic neutralization of the compound, and in rare cases, a lethal cytokine storm. One critical factor that can induce or contribute to an anti-drug antibody (ADA) response is believed to be the presence of aggregated proteins in protein-based therapeutics. There is a high level of variability in the aggregation of different proteins, which adds to the complexity in understanding the immune response to these drugs. Furthermore, the level of glycosylation of proteins, which increases drug stability, functionality, and serum half-life, is highly variable and may influence their immunogenicity. Considering the abundance of literature on the effect of aggregation and glycosylation on the immunogenicity of protein-based therapeutics, this review aims to summarize the current knowledge and clarify the immunogenic effects of different protein-based therapeutics such as mAbs. This review focuses on the properties of aggregated proteins and elucidates their relationship with immunogenicity. The contribution of different immune cell subsets and the mechanisms in aggregation-induced immunogenicity are also reviewed. Finally, the potential effects of each glycan, such as sialic acid, mannose, and fucose, on protein-based therapeutics' immunogenicity and stability is discussed.
Collapse
Affiliation(s)
- Sina Sarsarshahi
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Sanghati Bhattacharya
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Zeb R Zacharias
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Human Immunology Core, University of Iowa, Iowa City, IA, United States
| | - Eman S Kamel
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Jon C D Houtman
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Human Immunology Core, University of Iowa, Iowa City, IA, United States; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Reza Nejadnik
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States.
| |
Collapse
|
15
|
Lauridsen KM, Møller HJ, Kristensen MW, Fristrup N, Donskov F, Hokland M, Andersen MN. Soluble CD206 in metastatic renal cell carcinoma: Relation to clinical-biochemical parameters and patient outcome. Int J Cancer 2025; 156:875-885. [PMID: 39319597 DOI: 10.1002/ijc.35194] [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: 04/13/2024] [Revised: 08/11/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024]
Abstract
The mannose receptor (MR/CD206) is a marker of M2-like tumor-associated macrophages. Membrane CD206 can be shed, releasing the receptor as a soluble protein (sCD206), which can be measured in serum. Here, we investigated the biomarker potential of sCD206 in patients with metastatic renal cell carcinoma (mRCC). Serum sCD206 was measured by an enzyme-linked immunosorbent assay in 88 mRCC patients and 20 healthy controls (HCs). At diagnosis, serum sCD206 was elevated in patients with intermediate-risk mRCC according to the Memorial Sloan Kettering Cancer Center (MSKCC) risk score, compared to both HCs and patients with favorable MSKCC risk score. Furthermore, sCD206 levels correlated with both sCD163 and C-reactive protein. Soluble CD206 levels decreased after treatment initiation (p < .0001 at 5 weeks) but with a tendency toward elevated levels at time of progression, compared to baseline (p = .06). In univariate survival analysis, high levels of serum sCD206 at baseline was a significant risk factor associated with reduced overall survival (hazard ratio [HR] = 1.37, 95% confidence interval: 1.12-1.67, p = .002). Stratified by clinical risk scores, increased sCD206 was still a statistically significant risk factor of overall mortality (p < .01) in the intermediate-risk group by both the MSKCC (HR = 1.48) and the newer International Metastatic RCC Database Consortium (IMDC) score (HR = 1.53). Furthermore, addition of sCD206 as a dichotomized variable to the IMDC risk score enabled separation of the intermediate-risk group into two groups with survival comparable to those with favorable and poor risk, respectively. Overall, sCD206 is a potential add-on biomarker for mRCC patients in the intermediate-risk group of the current clinical risk scores.
Collapse
Affiliation(s)
- Kasper Munch Lauridsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Holger Jon Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mie Wolff Kristensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Fristrup
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Frede Donskov
- Department of Oncology, University Hospital of Southern Denmark, Esbjerg, Denmark
| | | | - Morten Nørgaard Andersen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
16
|
Weerachatyanukul W, Pooljun C, Chotwiwatthanakun C, Jariyapong P. Binding of Infectious Hypodermal and Haematopoietic Necrosis Virus-Like Particles to Mannose Receptor Stimulates Antimicrobial Responses in Immune-Related Tissues of Peneaus vannamei. JOURNAL OF FISH DISEASES 2025; 48:e14051. [PMID: 39609717 DOI: 10.1111/jfd.14051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/02/2024] [Accepted: 11/08/2024] [Indexed: 11/30/2024]
Abstract
Mannose receptor (MR) is a transmembrane protein and a type of pattern-recognition receptor (PRR) that plays a critical role in the immunity of mammals and fish. In this study, we examined the role of MR in binding with infectious hypodermal and haematopoietic necrosis virus-like particle (IHHN-VLP) and the downstream immune pathway that it triggers in the shrimp Peneaus vannamei. Upon IHHN-VLP challenge, transcripts of MR in P. vannamei (PvMR) increased significantly in all examined tissues, particularly those related to shrimp immunity, including hemocyte, hepatopancreas and gill tissues. Specifically, IHHN-VLP bound to the 34-kDa PvMR protein in shrimp-tissue extracts. Immunohistochemistry results of hemocytes showed that PvMR was initially localised on the plasma membrane but later internalised and dispersed throughout the cytoplasm after IHHN-VLP administration. Binding between IHHN-VLP and PvMR also induced significant upregulation of genes for the antimicrobial peptides (AMPs) penaeidin 3 and crustin, presumably to protect the shrimp against the viral infection. However, knocking down PvMR resulted in down-regulation of all immune-related genes examined. Overall, as an immune-related PRR, PvMR serves as a receptor for invading viruses, which then trigger the expression of AMPs. Strategic designs using PvMR could be developed to either block the interaction of native virus with the host cells or provoke its up-regulation to enhance shrimp immunity, which could open up opportunities to fight against IHHNV infection in shrimp.
Collapse
Affiliation(s)
| | - Chettupon Pooljun
- Akkhraratchakumari Veterinary College, Walailak University, Nakhonsrithammarat, Thailand
| | | | | |
Collapse
|
17
|
Wang Z, Kushibiki H, Tarusawa T, Osonoi S, Ogasawara S, Miura C, Sasaki T, Ryuzaki M, Yagihashi S, Mizukami H. Hypertension is associated with the reduction in epidermal small fibres independently of sural nerve inflammation in type 2 diabetic subjects. J Neurochem 2025; 169:e16235. [PMID: 39453752 PMCID: PMC11808456 DOI: 10.1111/jnc.16235] [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: 08/08/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/27/2024]
Abstract
Diabetic polyneuropathy (DPN) is a multifactorial disease associated not only with hyperglycaemia but also with circulatory disturbances such as hypertension. A close interaction between the immune system and hypertension is known. It remains unclear whether the inflammatory response is associated with hypertension in the pathology of human DPN. Autopsied patients were evaluated: 7 non-diabetic patients (nDM), 11 non-diabetic patients with hypertension (nDMHT), 6 patients with diabetes (DM) and 9 patients with hypertension and diabetes (DMHT). Intraepidermal nerve fibre density (IENFD) was examined by immunofluorescent staining. Dissected sural nerve (SNs) were morphometrically quantified. Dermal and endoneurial macrophage infiltration was evaluated by double immunostaining using anti-CD68 and anti-CD206 antibodies. IENFD was significantly decreased in DM compared to nDM (p < 0.05) and was further decreased in DMHT (p < 0.05). Myelinated nerve fibre density (MNFD) in the SN was significantly decreased in DM compared with nDM (p < 0.05) and further decreased in DMHT (p < 0.01 vs. DM). The infiltration of CD206-/CD68+ proinflammatory macrophages in the SN was significantly increased in DM compared to nDM (p < 0.05), whilst the number of CD206+/CD68+ anti-inflammatory macrophages was decreased in DM (p < 0.05). Hypertension had no impact on macrophage infiltration. The ratio of CD206- and CD206+ macrophage was negatively correlated with MNFD (r = 0.42, p < 0.05) but not IENFD (r = 0.30, p = 0.09). Dermal CD206+ macrophage infiltration was similar amongst all groups. Diabetes complicated by hypertension significantly increased the total diffusion barrier thickness (p < 0.01 vs. DM). Total diffusion barrier thickness was inversely correlated with both IENFD (r = -0.59, p < 0.01) and MNFD (r =-0.62, p < 0.01). Our results suggest that vascular factors and inflammation might be synergistically involved in pathological changes in human diabetic patients through different mechanisms.
Collapse
Affiliation(s)
- Zhenchao Wang
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| | - Hanae Kushibiki
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| | - Takefusa Tarusawa
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| | - Sho Osonoi
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| | - Saori Ogasawara
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| | - Chinatsu Miura
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| | - Takanori Sasaki
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| | - Masaki Ryuzaki
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| | - Soroku Yagihashi
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiJapan
| |
Collapse
|
18
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
19
|
Tian JH, Huang S, Wang ZH, Li JJ, Song X, Jiang ZT, Shi BS, Zhao YY, Zhang HY, Wang KR, Hu XY, Zhang X, Guo DS. Supramolecular discrimination and diagnosis-guided treatment of intracellular bacteria. Nat Commun 2025; 16:1016. [PMID: 39863571 PMCID: PMC11762306 DOI: 10.1038/s41467-025-56308-9] [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: 06/28/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Pathogenic intracellular bacteria pose a significant threat to global public health due to the barriers presented by host cells hindering the timely detection of hidden bacteria and the effective delivery of therapeutic agents. To address these challenges, we propose a tandem diagnosis-guided treatment paradigm. A supramolecular sensor array is developed for simple, rapid, accurate, and high-throughput identification of intracellular bacteria. This diagnostic approach executes the significant guiding missions of screening a customized host-guest drug delivery system by disclosing the rationale behind the discrimination. We design eight azocalix[4]arenes with differential active targeting, cellular internalization, and hypoxia responsiveness to penetrate cells and interact with bacteria. Loaded with fluorescent indicators, these azocalix[4]arenes form a sensor array capable of discriminating eight intracellular bacterial species without cell lysis or separation. By fingerprinting specimens collected from bacteria-infected mice, the facilitated accurate diagnosis offers valuable guidance for selecting appropriate antibiotics. Moreover, mannose-modified azocalix[4]arene (ManAC4A) is screened as a drug carrier efficiently taken up by macrophages. Doxycycline loaded with ManAC4A exhibits improved efficacy against methicillin-resistant Staphylococcus aureus-infected peritonitis. This study introduces an emerging paradigm to intracellular bacterial diagnosis and treatment, offering broad potential in combating bacterial infectious diseases.
Collapse
Affiliation(s)
- Jia-Hong Tian
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Siyuan Huang
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Han Wang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Juan-Juan Li
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Xianhui Song
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Tao Jiang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Bing-Sen Shi
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ying-Ying Zhao
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Hui-Yan Zhang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ke-Rang Wang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Xin-Yue Hu
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
| | - Xinge Zhang
- College of Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China.
| | - Dong-Sheng Guo
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, Kashi University, Kashi, China.
- College of Chemistry and Environmental Sciences, Kashi University, Kashi, China.
| |
Collapse
|
20
|
Wollman J, Wanniarachchi K, Pradhan B, Huang L, Kerkvliet JG, Hoppe AD, Thiex NW. Mannose receptor (MRC1) mediates uptake of dextran by bone marrow-derived macrophages. Mol Biol Cell 2024; 35:ar153. [PMID: 39504444 PMCID: PMC11656472 DOI: 10.1091/mbc.e24-08-0355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024] Open
Abstract
Macrophages survey their environment using receptor-mediated endocytosis and pinocytosis. Receptor-mediated endocytosis allows internalization of specific ligands, whereas pinocytosis nonselectively internalizes extracellular fluids and solutes. CRISPR/Cas9 whole-genome screens were used to identify genes regulating constitutive and growth factor-stimulated dextran uptake in murine bone marrow-derived macrophages (BMDM). The mannose receptor c-type 1 (MRC1/CD206) was a top hit in the screen. Targeted gene disruptions of Mrc1 reduced dextran uptake but had little effect on fluid-phase uptake of Lucifer yellow. Other screen hits also differentially affected the uptake of dextran and Lucifer yellow, indicating internalization by separate mechanisms. Visualization of dextran and Lucifer yellow uptake by microscopy showed enrichment of dextran in small puncta, which was inhibitable by mannan, a ligand of MRC1. In contrast, Lucifer yellow predominantly was internalized in larger macropinosomes. In addition, IL4-treated BMDMs internalized more dextran than untreated BMDM correlating with increased MRC1 expression. Therefore, dextran is not an effective marker for pinocytosis in BMDMs since it is internalized by receptor-mediated process. Numerous genes that regulate dextran internalization in primary murine macrophages were identified in the whole-genome screens, which can inform understanding of the regulation of MRC1 expression and MRC1-mediated uptake in macrophages.
Collapse
Affiliation(s)
- Jared Wollman
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| | - Kevin Wanniarachchi
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| | - Bijaya Pradhan
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| | - Lu Huang
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| | - Jason G Kerkvliet
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
- Chemistry, Biochemistry and Physics Department, South Dakota State University, Brookings, SD 57007
| | - Adam D Hoppe
- Chemistry, Biochemistry and Physics Department, South Dakota State University, Brookings, SD 57007
| | - Natalie W Thiex
- Biology and Microbiology Department, South Dakota State University, Brookings, SD 57007
| |
Collapse
|
21
|
Domenici G, Lopes NF, Trindade G, Ramella Gal I, Miret Minard J, Rebelo SP, Freitas C, Duarte N, Brito C. Assessing Novel Antibody-Based Therapies in Reconstructive 3D Cell Models of the Tumor Microenvironment. Adv Biol (Weinh) 2024; 8:e2400431. [PMID: 39601467 DOI: 10.1002/adbi.202400431] [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: 07/23/2024] [Revised: 10/21/2024] [Indexed: 11/29/2024]
Abstract
Targeted, combinatorial, and immunomodulatory therapies, such as antibody-drug conjugates (ADCs) and immunomodulatory antibodies (Abs), are powerful weapons against tumor cells and immune cells within the tumor microenvironment (TME). Therefore, the evaluation of such therapies should be conducted in pre-clinical models able to recapitulate the complex cellular and molecular crosstalk of the TME. To build-in critical hallmarks of the TME, a breast cancer heterotypic 3D cell model (3D-3) is devised using a microencapsulation strategy with an inert biomaterial (alginate) and agitation-based cultures. Both stromal and immune components are added to multicellular tumor spheroids, therefore fostering cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) immunomodulatory interactions. The potential of the methodology to assess Ab-based therapies is then addressed by employing a series of anti-HER2-based ADCs. ADCs induced tumor-cell specific cytotoxicity toward HER2+ breast cancer spheroids while sparing HER2-negative CAFs. In addition, an immunomodulatory blocking Ab against colony-stimulating factor 1 receptor (CSF1R) decreases the expression of immunosuppressive and anti-inflammatory markers in TAMs, like what is previously observed upon in vivo α-CSF1R administration. Collectively, the human TME-based 3D-3 cell model is a suitable tool to evaluate the anti-tumor and immunomodulatory potential of novel antibody-based therapies directed against TME targets, such as cancer cells and macrophages.
Collapse
Affiliation(s)
- Giacomo Domenici
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Nuno F Lopes
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Gonçalo Trindade
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Isabella Ramella Gal
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Joan Miret Minard
- Department of Chemical, Biological and Environmental Engineering, Universitat Autònoma de Barcelona, Plaça Cívica, Bellaterra, 08193, Spain
| | - Sofia P Rebelo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
| | - Catarina Freitas
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Nádia Duarte
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, 2780-901, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, 2780-157, Portugal
| |
Collapse
|
22
|
Xu X, Miao C, Yang S, Xiao L, Gao Y, Wu F, Xu J. Investigating potential drug targets for IgA nephropathy and membranous nephropathy through multi-queue plasma protein analysis: a Mendelian randomization study based on SMR and co-localization analysis. BioData Min 2024; 17:49. [PMID: 39516845 PMCID: PMC11545554 DOI: 10.1186/s13040-024-00405-w] [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: 07/27/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Membranous nephropathy (MN) and IgA nephropathy (IgAN) pose challenges in clinical treatment with existing therapies primarily focusing on symptom relief and often yielding unsatisfactory outcomes. The search for novel drug targets remains crucial to address the shortcomings in managing both kidney diseases. METHODS Utilizing GWAS data for MN (ncase = 2150, ncontrol = 5829) and IgAN (ncase = 15587, ncontrol = 462197), instrumental variables for plasma proteins were derived from recent GWAS. Sensitivity analysis involved bidirectional Mendelian randomization analysis, MR Steiger, Bayesian co-localization, and Phenotype scanning. The SMR analysis using eQTL data from the eQTLGen Consortium was conducted to assess the availability of selected protein targets. The PPI network was constructed to reveal potential associations with existing drug treatment targets. RESULTS The study, subjected to the stringent Bonferroni correction, revealed significant associations: four proteins with MN and three proteins with IgAN. In plasma protein cis-pQTL data from two cohorts, an increase in one standard deviation in PLA2R1 (OR = 2.01, 95%CI = 1.83-2.21), AIF1 (OR = 9.04, 95%CI = 4.69-17.41), MLN (OR = 3.79, 95%CI = 2.12-6.78), and NFKB1 (OR = 29.43, 95%CI = 7.73-112.0) was associated with an increased risk of MN. Additionally, in plasma protein cis-pQTL data, a standard deviation increase in FCGR3B (OR = 1.15, 95%CI = 1.09-1.22) and BTN3A1 (OR = 4.05, 95%CI = 2.65-6.19) correlated with elevated IgAN risk, while AIF1 (OR = 0.58, 95%CI = 0.46-0.73) exhibited IgAN protection. Bayesian co-localization indicated that PLA2R1 (coloc.abf-PPH4 = 0.695), NFKB1 (coloc.abf-PPH4 = 0.949), FCGR3B (coloc.abf-PPH4 = 0.909), and BTN3A1 (coloc.abf-PPH4 = 0.685) share the same variants associated with MN and IgAN. The SMR analysis indicated a causal link between NFKB1 and BTN3A1 plasma protein eQTL in both conditions, and BTN3A1 was validated externally. CONCLUSION Genetically influenced plasma levels of PLA2R1 and NFKB1 impact MN risk, while FCGR3B and BTN3A1 levels are causally linked to IgAN risk, suggesting potential drug targets for further clinical exploration, notably BTN3A1 for IgAN.
Collapse
Affiliation(s)
- Xinyi Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Changhong Miao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shirui Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lu Xiao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Ying Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fangying Wu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jianbo Xu
- Department of Clinical Laboratory, Jinhua Maternal and Child Health Care Hospital, Jinhua, Zhejiang, 321000, China.
| |
Collapse
|
23
|
Colaço M, Cruz MT, de Almeida LP, Borges O. Mannose and Lactobionic Acid in Nasal Vaccination: Enhancing Antigen Delivery via C-Type Lectin Receptors. Pharmaceutics 2024; 16:1308. [PMID: 39458637 PMCID: PMC11510408 DOI: 10.3390/pharmaceutics16101308] [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: 08/20/2024] [Revised: 09/24/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Nasal vaccines are a promising strategy for enhancing mucosal immune responses and preventing diseases at mucosal sites by stimulating the secretion of secretory IgA, which is crucial for early pathogen neutralization. However, designing effective nasal vaccines is challenging due to the complex immunological mechanisms in the nasal mucosa, which must balance protection and tolerance against constant exposure to inhaled pathogens. The nasal route also presents unique formulation and delivery hurdles, such as the mucous layer hindering antigen penetration and immune cell access. METHODS This review focuses on cutting-edge approaches to enhance nasal vaccine delivery, particularly those targeting C-type lectin receptors (CLRs) like the mannose receptor and macrophage galactose-type lectin (MGL) receptor. It elucidates the roles of these receptors in antigen recognition and uptake by antigen-presenting cells (APCs), providing insights into optimizing vaccine delivery. RESULTS While a comprehensive examination of targeted glycoconjugate vaccine development is outside the scope of this study, we provide key examples of glycan-based ligands, such as lactobionic acid and mannose, which can selectively target CLRs in the nasal mucosa. CONCLUSIONS With the rise of new viral infections, this review aims to facilitate the design of innovative vaccines and equip researchers, clinicians, and vaccine developers with the knowledge to enhance immune defenses against respiratory pathogens, ultimately protecting public health.
Collapse
Affiliation(s)
- Mariana Colaço
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria T. Cruz
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Olga Borges
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
24
|
Cao Q, Li J, Chen M. Bioinformatics analysis of neutrophil-associated hub genes and ceRNA network construction in septic cardiomyopathy. Aging (Albany NY) 2024; 16:12833-12849. [PMID: 39216003 PMCID: PMC11501391 DOI: 10.18632/aging.206092] [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: 03/11/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024]
Abstract
Septic cardiomyopathy (SCM) is a critical sepsis complication characterized by reversible cardiac depression during early septic shock. Neutrophils, integral to innate immunity, can mediate organ damage when abnormal, but their specific role in sepsis-induced myocardial damage remains elusive. Our study focuses on elucidating the role of Neutrophil-Related Genes (NRGs) in SCM, finding early diagnosis and treatment biomarkers. We identified shared differentially expressed genes (DEGs) from datasets GSE79962 and GSE44363 and pinpointed hub DEGs using the cytoHubba plugin in Cytoscape software. The Neutrophil-Related Hub Gene (NRHG) MRC1 was identified via intersecting hub DEGs with NRGs from WGCNA. We validated MRC1's abnormal expression in SCM using our data and external datasets. Furthermore, a neutrophil-related ceRNA network (AC145207.5/ miR-23a-3p/MRC1) was constructed and validated. Our findings reveal MRC1 as a potential NRHG in SCM pathogenesis, offering insights into neutrophil-mediated mechanisms in SCM and providing a novel molecular target for early diagnosis and intervention in SCM.
Collapse
Affiliation(s)
- Qingfei Cao
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jing Li
- Department of Pediatric, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Meixue Chen
- Department of Pediatric, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
25
|
Soni S, Kim KM, Sarkar B, Rodell CB. Uptake of Cyclodextrin Nanoparticles by Macrophages is Dependent on Particle Size and Receptor-Mediated Interactions. ACS APPLIED BIO MATERIALS 2024; 7:4856-4866. [PMID: 38231485 PMCID: PMC11252246 DOI: 10.1021/acsabm.3c00985] [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: 10/22/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/18/2024]
Abstract
Physiochemical properties of nanoparticles, such as their size and chemical composition, dictate their interaction with professional phagocytes of the innate immune system. Macrophages, in particular, are key regulators of the immune microenvironment that heavily influence particle biodistribution as a result of their uptake. This attribute enables macrophage-targeted delivery, including for phenotypic modulation. Saccharide-based materials, including polyglucose polymers and nanoparticles, are efficient vehicles for macrophage-targeted delivery. Here, we investigate the influence of particle size on cyclodextrin nanoparticle (CDNP) uptake by macrophages and further examine the receptor-mediated interactions that drive macrophage-targeted delivery. We designed and synthesized CDNPs ranging in size from 25 nm to >100 nm in diameter. Increasing particle size was correlated with greater uptake by macrophages in vitro. Both scavenger receptor A1 and mannose receptor were critical mediators of macrophage-targeted delivery, inhibition of which reduced the extent of uptake. Finally, we investigated the cellular bioavailability of drug-loaded CDNPs using a model anti-inflammatory drug, celastrol, which demonstrated that drug bioactivity is improved by CDNP loading relative to free drug alone. This study thus elucidates the interactions between the polyglucose nanoparticles and macrophages, thereby facilitating their application in macrophage-targeted drug delivery that has applications in the context of tissue injury and repair.
Collapse
Affiliation(s)
- Shreya
S. Soni
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Kenneth M. Kim
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
- Department
of Microbiology and Immunology, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Biplab Sarkar
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Christopher B. Rodell
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
26
|
Hwang M, Bergmann CC. Neurotropic murine coronavirus mediated demyelination: Factors dampening pathogenesis. J Neuroimmunol 2024; 393:578382. [PMID: 38850674 DOI: 10.1016/j.jneuroim.2024.578382] [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: 03/31/2024] [Revised: 05/12/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Virus infections and autoimmune responses are implicated as primary triggers of demyelinating diseases. Specifically, the association of Epstein-Barr virus (EBV) infection with development of multiple sclerosis (MS) has re-ignited an interest in virus induced autoimmune responses to CNS antigens. Nevertheless, demyelination may also be caused by immune mediated bystander pathology in an attempt to control direct infection in the CNS. Tissue damage as a result of anti-viral responses or low level viral persistence may lead to immune activation manifesting in demyelinating lesions, axonal damage and clinical symptoms. This review focuses on the neurotropic mouse coronavirus induced demyelination model to highlight how immune responses activated during the acute phase pave the way to dampen pathology and promote repair. We specifically discuss the role of immune dampening factors programmed cell death ligand 1 (PD-L1) and interleukin (IL)-10, as well as microglia and triggering receptor expressed on myeloid cells 2 (Trem2), in limiting demyelination independent of viral persistence.
Collapse
Affiliation(s)
- Mihyun Hwang
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Cornelia C Bergmann
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
27
|
Snyder Y, Mann FAT, Middleton J, Murashita T, Carney J, Bianco RW, Jana S. Non-immune factors cause prolonged myofibroblast phenotype in implanted synthetic heart valve scaffolds. APPLIED MATERIALS TODAY 2024; 39:102323. [PMID: 39131741 PMCID: PMC11308761 DOI: 10.1016/j.apmt.2024.102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The clinical application of heart valve scaffolds is hindered by complications associated with the activation of valvular interstitial cell-like (VIC-like) cells and their transdifferentiation into myofibroblasts. This study aimed to examine several molecular pathway(s) that may trigger the overactive myofibroblast phenotypes in the implanted scaffolds. So, we investigated the influence of three molecular pathways - macrophage-induced inflammation, the TGF-β1-SMAD2, and WNT/β-catenin β on VIC-like cells during tissue engineering of heart valve scaffolds. We implanted electrospun heart valve scaffolds in adult sheep for up to 6 months in the right ventricular outflow tract (RVOT) and analyzed biomolecular (gene and protein) expression associated with the above three pathways by the scaffold infiltrating cells. The results showed a gradual increase in gene and protein expression of markers related to the activation of VIC-like cells and the myofibroblast phenotypes over 6 months of scaffold implantation. Conversely, there was a gradual increase in macrophage activity for the first three months after scaffold implantation. However, a decrease in macrophage activity from three to six months of scaffold tissue engineering suggested that immunological signal factors were not the primary cause of myofibroblast phenotype. Similarly, the gene and protein expression of factors associated with the TGF-β1-SMAD2 pathway in the cells increased in the first three months but declined in the next three months. Contrastingly, the gene and protein expression of factors associated with the WNT/β-catenin pathway increased significantly over the six-month study. Thus, the WNT/β-catenin pathway could be the predominant mechanism in activating VIC-like cells and subsequent myofibroblast phenotype.
Collapse
Affiliation(s)
- Yuriy Snyder
- Department of Chemical and Biomedical Engineering, University of Missouri, 1406 Rollins Street, Columbia, MO 65211, USA
| | - FA Tony Mann
- Veterinary Health Center, University of Missouri, 900 East Campus Drive, Columbia, MO 65211-0001
| | - John Middleton
- Veterinary Health Center, University of Missouri, 900 East Campus Drive, Columbia, MO 65211-0001
| | - Takashi Murashita
- Department of Surgery, School of Medicine, University of Missouri, One Hospital Drive, Columbia, MO 65212
| | - John Carney
- Experimental Surgical Services, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455
| | - Richard W. Bianco
- Experimental Surgical Services, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455
| | - Soumen Jana
- Department of Chemical and Biomedical Engineering, University of Missouri, 1406 Rollins Street, Columbia, MO 65211, USA
| |
Collapse
|
28
|
Rojekar S, Gholap AD, Togre N, Bhoj P, Haeck C, Hatvate N, Singh N, Vitore J, Dhoble S, Kashid S, Patravale V. Current status of mannose receptor-targeted drug delivery for improved anti-HIV therapy. J Control Release 2024; 372:494-521. [PMID: 38849091 DOI: 10.1016/j.jconrel.2024.06.002] [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: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
In the pursuit of achieving better therapeutic outcomes in the treatment of HIV, innovative drug delivery strategies have been extensively explored. Mannose receptors, which are primarily found on macrophages and dendritic cells, offer promising targets for drug delivery due to their involvement in HIV pathogenesis. This review article comprehensively evaluates recent drug delivery system advancements targeting the mannose receptor. We have systematically described recent developments in creating and utilizing drug delivery platforms, including nanoparticles, liposomes, micelles, noisomes, dendrimers, and other nanocarrier systems targeted at the mannose receptor. These strategies aim to enhance drug delivery specificity, bioavailability, and therapeutic efficacy while decreasing off-target effects and systemic toxicity. Furthermore, the article delves into how mannose receptors and HIV interact, highlighting the potential for exploiting this interaction to enhance drug delivery to infected cells. The review covers essential topics, such as the rational design of nanocarriers for mannose receptor recognition, the impact of physicochemical properties on drug delivery performance, and how targeted delivery affects the pharmacokinetics and pharmacodynamics of anti-HIV agents. The challenges of these novel strategies, including immunogenicity, stability, and scalability, and future research directions in this rapidly growing area are discussed. The knowledge synthesis presented in this review underscores the potential of mannose receptor-based targeted drug delivery as a promising avenue for advancing HIV treatment. By leveraging the unique properties of mannose receptors, researchers can design drug delivery systems that cater to individual needs, overcome existing limitations, and create more effective and patient-friendly treatments in the ongoing fight against HIV/AIDS.
Collapse
Affiliation(s)
- Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Namdev Togre
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Priyanka Bhoj
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Clement Haeck
- Population Council, , Center for Biomedical Research, 1230 York Avenue, New York, NY 10065, USA
| | - Navnath Hatvate
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Nidhi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India
| | - Jyotsna Vitore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Sagar Dhoble
- Department of Pharmacology and Toxicology, R. K. Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Snehal Kashid
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
29
|
Korpidou M, Becker J, Tarvirdipour S, Dinu IA, Becer CR, Palivan CG. Glycooligomer-Functionalized Catalytic Nanocompartments Co-Loaded with Enzymes Support Parallel Reactions and Promote Cell Internalization. Biomacromolecules 2024; 25:4492-4509. [PMID: 38910355 PMCID: PMC11238334 DOI: 10.1021/acs.biomac.4c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024]
Abstract
A major shortcoming associated with the application of enzymes in drug synergism originates from the lack of site-specific, multifunctional nanomedicine. This study introduces catalytic nanocompartments (CNCs) made of a mixture of PDMS-b-PMOXA diblock copolymers, decorated with glycooligomer tethers comprising eight mannose-containing repeating units and coencapsulating two enzymes, providing multifunctionality by their in situ parallel reactions. Beta-glucuronidase (GUS) serves for local reactivation of the drug hymecromone, while glucose oxidase (GOx) induces cell starvation through glucose depletion and generation of the cytotoxic H2O2. The insertion of the pore-forming peptide, melittin, facilitates diffusion of substrates and products through the membranes. Increased cell-specific internalization of the CNCs results in a substantial decrease in HepG2 cell viability after 24 h, attributed to simultaneous production of hymecromone and H2O2. Such parallel enzymatic reactions taking place in nanocompartments pave the way to achieve efficient combinatorial cancer therapy by enabling localized drug production along with reactive oxygen species (ROS) elevation.
Collapse
Affiliation(s)
- Maria Korpidou
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
| | - Jonas Becker
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Shabnam Tarvirdipour
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
| | - Ionel Adrian Dinu
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
| | - C. Remzi Becer
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Cornelia G. Palivan
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
- NCCR
Molecular Systems Engineering, Mattenstrasse 22, Basel 4002, Switzerland
| |
Collapse
|
30
|
Andriana P, Fair-Mäkelä R, Liljenbäck H, Kärnä S, Iqbal I, Makrypidi K, Rajander J, Pirmettis I, Li XG, Jalkanen S, Saraste A, Salmi M, Roivainen A. Macrophage mannose receptor CD206 targeting of fluoride-18 labeled mannosylated dextran: A validation study in mice. Eur J Nucl Med Mol Imaging 2024; 51:2216-2228. [PMID: 38532026 PMCID: PMC11178572 DOI: 10.1007/s00259-024-06686-x] [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: 01/12/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024]
Abstract
PURPOSE Aluminum fluoride-18-labeled 1,4,7-triazacyclononane-1,4,7-triacetic acid-conjugated mannosylated dextran derivative (Al[18F]F-NOTA-D10CM) is a new tracer for PET imaging. We report here on in vitro and in vivo validation of the tracer's ability to target the macrophage mannose receptor CD206. METHODS First, the uptake of intravenously (i.v.) administered Al[18F]F-NOTA-D10CM was compared between wild-type (WT) and CD206-/- knockout (KO) mice. C57BL/6N mice were injected with complete Freund's adjuvant (CFA) in the left hind leg and the uptake of Al[18F]F-NOTA-D10CM after i.v. or intradermal (i.d.) injection was studied at 5 and 14 days after CFA induction of inflammation. Healthy C57BL/6N mice were studied as controls. Mice underwent PET/CT on consecutive days with [18F]FDG, i.v. Al[18F]F-NOTA-D10CM, and i.d. Al[18F]F-NOTA-D10CM. After the last imaging, Al[18F]F-NOTA-D10CM was i.v. injected for an ex vivo biodistribution study and autoradiography of inflamed tissues. Blood plasma samples were analyzed using high-performance liquid chromatography. To evaluate the specificity of Al[18F]F-NOTA-D10CM binding, an in vitro competitive displacement study was performed on inflamed tissue sections using autoradiography. CD206 expression was assessed by immunohistochemical staining. RESULTS Compared with WT mice, the uptake of Al[18F]F-NOTA-D10CM was significantly lower in several CD206-/- KO mice tissues, including liver (SUV 8.21 ± 2.51 vs. 1.06 ± 0.16, P < 0.001) and bone marrow (SUV 1.63 ± 0.37 vs. 0.22 ± 0.05, P < 0.0001). The uptake of i.v. injected Al[18F]F-NOTA-D10CM was significantly higher in inflamed ankle joint (SUV 0.48 ± 0.13 vs. 0.18 ± 0.05, P < 0.0001) and inflamed foot pad skin (SUV 0.41 ± 0.10 vs. 0.04 ± 0.01, P < 0.0001) than in the corresponding tissues in healthy mice. The i.d.-injected Al[18F]F-NOTA-D10CM revealed differences between CFA-induced lymph node activation and lymph nodes in healthy mice. Ex vivo γ-counting, autoradiography, and immunohistochemistry supported the results, and a decrease of ~ 80% in the binding of Al[18F]F-NOTA-D10CM in the displacement study with excess NOTA-D10CM confirmed that tracer binding was specific. At 60 min after i.v. injection, an average 96.70% of plasma radioactivity was derived from intact Al[18F]F-NOTA-D10CM, indicating good in vivo stability. The uptake of Al[18F]F-NOTA-D10CM into inflamed tissues was positively associated with the area percentage of CD206-positive staining. CONCLUSION The uptake of mannosylated dextran derivative Al[18F]F-NOTA-D10CM correlated with CD206 expression and the tracer appears promising for inflammation imaging.
Collapse
Affiliation(s)
- Putri Andriana
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Ruth Fair-Mäkelä
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- Turku Center of Disease Modeling, University of Turku, Turku, Finland
| | - Salli Kärnä
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Imran Iqbal
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Konstantina Makrypidi
- Institute of Nuclear and Radiological Science and Technology, Energy and Safety, NCSR "Demokritos", Athens, Greece
| | - Johan Rajander
- Turku PET Centre, Accelerator Laboratory, Åbo Akademi University, Turku, Finland
| | - Ioannis Pirmettis
- Institute of Nuclear and Radiological Science and Technology, Energy and Safety, NCSR "Demokritos", Athens, Greece
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Department of Chemistry, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Marko Salmi
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Turku Center of Disease Modeling, University of Turku, Turku, Finland.
- Turku PET Centre, Turku University Hospital, Turku, Finland.
| |
Collapse
|
31
|
Niu L, Wang H, Luo G, Zhou J, Hu Z, Yan B. Advances in understanding immune homeostasis in latent tuberculosis infection. WIREs Mech Dis 2024; 16:e1643. [PMID: 38351551 DOI: 10.1002/wsbm.1643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 07/13/2024]
Abstract
Nearly one-fourth of the global population is infected by Mycobacterium tuberculosis (Mtb), and approximately 90%-95% remain asymptomatic as latent tuberculosis infection (LTBI), an estimated 5%-10% of those with latent infections will eventually progress to active tuberculosis (ATB). Although it is widely accepted that LTBI transitioning to ATB results from a disruption of host immune balance and a weakening of protective immune responses, the exact underlying immunological mechanisms that promote this conversion are not well characterized. Thus, it is difficult to accurately predict tuberculosis (TB) progression in advance, leaving the LTBI population as a significant threat to TB prevention and control. This article systematically explores three aspects related to the immunoregulatory mechanisms and translational research about LTBI: (1) the distinct immunocytological characteristics of LTBI and ATB, (2) LTBI diagnostic markers discovery related to host anti-TB immunity and metabolic pathways, and (3) vaccine development focus on LTBI. This article is categorized under: Infectious Diseases > Molecular and Cellular Physiology Infectious Diseases > Genetics/Genomics/Epigenetics Immune System Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Liangfei Niu
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| | - Hao Wang
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Geyang Luo
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| | - Jing Zhou
- Department of Pathology, Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| | - Zhidong Hu
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| | - Bo Yan
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
32
|
Dolla G, Nicolas S, Dos Santos LR, Bourgeois A, Pardossi-Piquard R, Bihl F, Zaghrini C, Justino J, Payré C, Mansuelle P, Garbers C, Ronco P, Checler F, Lambeau G, Petit-Paitel A. Ectodomain shedding of PLA2R1 is mediated by the metalloproteases ADAM10 and ADAM17. J Biol Chem 2024; 300:107480. [PMID: 38897568 PMCID: PMC11301074 DOI: 10.1016/j.jbc.2024.107480] [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: 12/03/2023] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Phospholipase A2 receptor 1 (PLA2R1) is a 180-kDa transmembrane protein that plays a role in inflammation and cancer and is the major autoantigen in membranous nephropathy, a rare but severe autoimmune kidney disease. A soluble form of PLA2R1 has been detected in mouse and human serum. It is likely produced by proteolytic shedding of membrane-bound PLA2R1 but the mechanism is unknown. Here, we show that human PLA2R1 is cleaved by A Disintegrin And Metalloprotease 10 (ADAM10) and ADAM17 in HEK293 cells, mouse embryonic fibroblasts, and human podocytes. By combining site-directed mutagenesis and sequencing, we determined the exact cleavage site within the extracellular juxtamembrane stalk of human PLA2R1. Orthologs and paralogs of PLA2R1 are also shed. By using pharmacological inhibitors and genetic approaches with RNA interference and knock-out cellular models, we identified a major role of ADAM10 in the constitutive shedding of PLA2R1 and a dual role of ADAM10 and ADAM17 in the stimulated shedding. We did not observe evidence for cleavage by β- or γ-secretase, suggesting that PLA2R1 may not be a substrate for regulated intramembrane proteolysis. PLA2R1 shedding occurs constitutively and can be triggered by the calcium ionophore ionomycin, the protein kinase C activator PMA, cytokines, and lipopolysaccharides, in vitro and in vivo. Altogether, our results show that PLA2R1 is a novel substrate for ADAM10 and ADAM17, producing a soluble form that is increased in inflammatory conditions and likely exerts various functions in physiological and pathophysiological conditions including inflammation, cancer, and membranous nephropathy.
Collapse
Affiliation(s)
- Guillaume Dolla
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Sarah Nicolas
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Ligia Ramos Dos Santos
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Laboratoire d'Excellence DistALZ, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Alexandre Bourgeois
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Laboratoire d'Excellence DistALZ, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Raphaëlle Pardossi-Piquard
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Laboratoire d'Excellence DistALZ, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Franck Bihl
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Christelle Zaghrini
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Joana Justino
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Christine Payré
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Pascal Mansuelle
- Plateforme de Protéomique de l'Institut de Microbiologie de la Méditerranée (IMM), Marseille Protéomique (MaP), Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS) FR3479, Marseille, France
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Pierre Ronco
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S1155, Paris, France; Sorbonne Université, Université Pierre et Marie Curie Paris 06, Paris, France
| | - Frédéric Checler
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Laboratoire d'Excellence DistALZ, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Gérard Lambeau
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France.
| | - Agnès Petit-Paitel
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France.
| |
Collapse
|
33
|
Liu Z, Liu B, Feng Y, Zhao L, Wang Q, He H, Yin T, Zhang Y, Yang L, Gou J, Tang X. Dual-Targeted Self-Adjuvant Heterocyclic Lipidoid@Polyester Hybrid Nanovaccines for Boosting Cancer Immunotherapy. ACS NANO 2024; 18:15557-15575. [PMID: 38837909 DOI: 10.1021/acsnano.4c00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Tumor vaccines have demonstrated a modest response rate, primarily attributed to their inefficient delivery to dendritic cells (DCs), low cross-presentation, DC-intrinsic immunosuppressive signals, and an immunosuppressive tumor microenvironment (TME). Here, draining lymph node (DLN)-targeted and tumor-targeted nanovaccines were proposed to address these limitations, and heterocyclic lipidoid (A18) and polyester (BR647) were synthesized to achieve dual-targeted cancer immunotherapy. Meanwhile, oligo hyaluronic acid (HA) and DMG-PEG2000-Mannose were incorporated to prepare dual-targeted nanovaccines encapsulated with STAT3 siRNA and model antigens. The nanovaccines were designed to target the DLN and the tumor, facilitating the delivery of cargo into the cytoplasm. These dual-targeted nanovaccines improved antigen presentation and DC maturation, activated the stimulator of interferon genes (STING) pathway, enhanced the pro-apoptotic effect, and stimulated antitumor immune responses. Additionally, these dual-targeted nanovaccines overcame immunosuppressive TME, reduced immunosuppressive cells, and promoted the polarization of tumor-associated neutrophils from N2 to N1. Among the four dual-targeted nanovaccines that induced robust antitumor responses, the heterocyclic lipidoid@polyester hybrid nanovaccines (MALO@HBNS) demonstrated the most promising results. Furthermore, a combination strategy involving MALO@HBNS and an anti-PD-L1 antibody exhibited an immensely powerful anticancer role. This work introduced a dual-targeted nanovaccine platform for antitumor treatment, suggesting its potential combination with an immune checkpoint blockade as a comprehensive anticancer strategy.
Collapse
Affiliation(s)
- Zixu Liu
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Boyuan Liu
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Yupeng Feng
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Linxuan Zhao
- Department of Pharmaceutics, College of Pharmacy Sciences, Jilin University, Changchun 130021, China
| | - Qingqing Wang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Haibing He
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Tian Yin
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Zhang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Li Yang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Jingxin Gou
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Xing Tang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang 110116, China
| |
Collapse
|
34
|
Wu Z, Huang C. Unveiling the Impact of MRC1 on Immune Infiltration and Patient's Prognosis: A Pan-Cancer Analysis Based on Single-Cell and Bulk Sequencing. Int J Gen Med 2024; 17:2575-2592. [PMID: 38855425 PMCID: PMC11162242 DOI: 10.2147/ijgm.s461144] [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: 01/24/2024] [Accepted: 05/20/2024] [Indexed: 06/11/2024] Open
Abstract
Purpose Mannose receptor C-type 1 (MRC1) is an endocytic lectin receptor primarily expressed in macrophages, dendritic cells, and some endothelial cells. However, the role of MRC1 in cancers remains unclear. Methods We analyzed MRC1 expression using The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), Cancer Cell Line Encyclopedia (CCLE), and single-cell datasets. We systematically explored the prognostic implications and diagnostic value of MRC1. Immune-related indicators, including immune cells, immune scores, and immune checkpoint molecules, were used to estimate their correlation with MRC1 expression. Finally, we explored its potential ties to immunotherapy success markers such as tumor mutation burden and DNA repair genes. Results MRC1 showed both pro- and anti-tumor leanings depending on the cancer types. High levels correlated with poorer outcomes in six cancers but improved prognosis in some cancers like glioblastoma multiforme. This trend extended to the immune arena, where MRC1 intertwined with diverse immune parameters, suggesting its influence on affecting the tumor's immunological landscape. Intriguingly, its expression positively associated with factors favoring immunotherapy efficacy while negatively correlating with some potential barriers. Single-cell analysis pinpointed a specific link between MRC1 and DNA damage/repair pathways in breast cancer. Conclusion Our study provides a comprehensive landscape of MRC1 levels and diverse regulatory patterns in different cancers, deepening the understanding of MRC1's roles in tumorigenesis and immunity.
Collapse
Affiliation(s)
- Zhiwei Wu
- Department of Organ Transplantation, XiangYa Hospital of Central South University, Changsha, People’s Republic of China
| | - Changhao Huang
- Department of Organ Transplantation, XiangYa Hospital of Central South University, Changsha, People’s Republic of China
| |
Collapse
|
35
|
Liu P, Fei L, Wu D, Zhang Z, Chen W, Li W, Yang Y. Progress in the metabolic kinetics and health benefits of functional polysaccharides from plants, animals and microbes: A review. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2024; 7:100526. [DOI: 10.1016/j.carpta.2024.100526] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
|
36
|
Nørregaard KS, Jürgensen HJ, Heltberg SS, Gårdsvoll H, Bugge TH, Schoof EM, Engelholm LH, Behrendt N. A proteomics-based survey reveals thrombospondin-4 as a ligand regulated by the mannose receptor in the injured lung. J Biol Chem 2024; 300:107284. [PMID: 38614208 PMCID: PMC11107221 DOI: 10.1016/j.jbc.2024.107284] [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: 02/09/2024] [Revised: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 04/15/2024] Open
Abstract
Receptor-mediated cellular uptake of specific ligands constitutes an important step in the dynamic regulation of individual protein levels in extracellular fluids. With a focus on the inflammatory lung, we here performed a proteomics-based search for novel ligands regulated by the mannose receptor (MR), a macrophage-expressed endocytic receptor. WT and MR-deficient mice were exposed to lipopolysaccharide, after which the protein content in their lung epithelial lining fluid was compared by tandem mass tag-based mass spectrometry. More than 1200 proteins were identified in the epithelial lining fluid using this unbiased approach, but only six showed a statistically different abundance. Among these, an unexpected potential new ligand, thrombospondin-4 (TSP-4), displayed a striking 17-fold increased abundance in the MR-deficient mice. Experiments using exogenous addition of TSP-4 to MR-transfected CHO cells or MR-positive alveolar macrophages confirmed that TSP-4 is a ligand for MR-dependent endocytosis. Similar studies revealed that the molecular interaction with TSP-4 depends on both the lectin activity and the fibronectin type-II domain of MR and that a closely related member of the TSP family, TSP-5, is also efficiently internalized by the receptor. This was unlike the other members of this protein family, including TSPs -1 and -2, which are ligands for a close MR homologue known as urokinase plasminogen activator receptor-associated protein. Our study shows that MR takes part in the regulation of TSP-4, an important inflammatory component in the injured lung, and that two closely related endocytic receptors, expressed on different cell types, undertake the selective endocytosis of distinct members of the TSP family.
Collapse
Affiliation(s)
- Kirstine S Nørregaard
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Henrik J Jürgensen
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Signe S Heltberg
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Henrik Gårdsvoll
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Erwin M Schoof
- Section for Protein Science and Biotherapeutics, Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lars H Engelholm
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Niels Behrendt
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
37
|
Dirand Z, Maraux M, Tissot M, Chatelain B, Supp D, Viennet C, Perruche S, Rolin G. Macrophage phenotype is determinant for fibrosis development in keloid disease. Matrix Biol 2024; 128:79-92. [PMID: 38485100 DOI: 10.1016/j.matbio.2024.03.001] [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: 08/16/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 04/02/2024]
Abstract
Keloid refers to a fibroproliferative disorder characterized by an accumulation of extracellular matrix (ECM) components at the dermis level, overgrowth beyond initial wound, and formation of tumor-like nodule areas. Treating keloid is still an unmet clinical need and the lack of an efficient therapy is clearly related to limited knowledge about keloid etiology, despite the growing interest of the scientific community in this pathology. In past decades, keloids were often studied in vitro through the sole prism of fibroblasts considered as the major effector of ECM deposition. Nevertheless, development of keloids results from cross-interactions of keloid fibroblasts (KFs) and their surrounding microenvironment, including immune cells such as macrophages. Our study aimed to evaluate the effect of M1 and M2 monocyte-derived macrophages on KFs in vitro. We focused on the effects of the macrophage secretome on fibrosis-related criteria in KFs, including proliferation, migration, differentiation, and ECM synthesis. First, we demonstrated that M2-like macrophages enhanced the fibrogenic profile of KFs in culture. Then, we surprisingly founded that M1-like macrophages can have an anti-fibrogenic effect on KFs, even in a pro-fibrotic environment. These results demonstrate, for the first time, that M1 and M2 macrophage subsets differentially impact the fibrotic fate of KFs in vitro, and suggest that restoring the M1/M2 balance to favor M1 in keloids could be an efficient therapeutic lever to prevent or treat keloid fibrosis.
Collapse
Affiliation(s)
- Zélie Dirand
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France
| | - Mélissa Maraux
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France
| | - Marion Tissot
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; DImaCell Imaging Resource Center, 25000 Besançon, France
| | - Brice Chatelain
- Service de Chirurgie Maxillo-faciale, Stomatologie et Odontologie Hospitalière, CHU Besançon, 25000 Besançon, France
| | - Dorothy Supp
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Scientific Staff, Shriners Children's Ohio, Dayton, Ohio, USA
| | - Céline Viennet
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; DImaCell Imaging Resource Center, 25000 Besançon, France
| | - Sylvain Perruche
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; MED'INN'Pharma 25000 Besançon, France
| | - Gwenaël Rolin
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; DImaCell Imaging Resource Center, 25000 Besançon, France; INSERM CIC-1431, CHU Besançon, 25000 Besançon, France.
| |
Collapse
|
38
|
Lei J, Qi S, Yu X, Gao X, Yang K, Zhang X, Cheng M, Bai B, Feng Y, Lu M, Wang Y, Li H, Yu G. Development of Mannosylated Lipid Nanoparticles for mRNA Cancer Vaccine with High Antigen Presentation Efficiency and Immunomodulatory Capability. Angew Chem Int Ed Engl 2024; 63:e202318515. [PMID: 38320193 DOI: 10.1002/anie.202318515] [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: 12/03/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/08/2024]
Abstract
Insufficient accumulation of lipid nanoparticles (LNPs)-based mRNA vaccines in antigen presenting cells remains a key barrier to eliciting potent antitumor immune responses. Herein, we develop dendritic cells (DCs) targeting LNPs by taking advantage of mannose receptor-mediated endocytosis. Efficient delivery of mRNA to DCs is achieved in vitro and in vivo utilizing the sweet LNPs (STLNPs-Man). Intramuscular injection of mRNA vaccine (STLNPs-Man@mRNAOVA ) results in a four-fold higher uptake by DCs in comparison with commercially used LNPs. Benefiting from its DCs targeting ability, STLNPs-Man@mRNAOVA significantly promotes the antitumor performances, showing a comparable therapeutic efficacy by using one-fifth of the injection dosage as the vaccine prepared from normal LNPs, thus remarkably avoiding the side effects brought by conventional mRNA vaccines. More intriguingly, STLNPs-Man@mRNAOVA exhibits the ability to downregulate the expression of cytotoxic T-lymphocyte-associated protein 4 on T cells due to the blockade of CD206/CD45 axis, showing brilliant potentials in promoting antitumor efficacy combined with immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Jiaqi Lei
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Shaolong Qi
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Xinyang Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Xiaomin Gao
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Kai Yang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Xueyan Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Meiqi Cheng
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Bing Bai
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Yunxuan Feng
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Meixin Lu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Yangfan Wang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
| | - Hongjian Li
- School of Medicine, Tsinghua University, 100084, Beijing, P. R. China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, 100084, Beijing, P. R. China
- School of Medicine, Tsinghua University, 100084, Beijing, P. R. China
| |
Collapse
|
39
|
Riemann B, Antoine T, Béduneau A, Pellequer Y, Lamprecht A, Moulari B. Active nanoparticle targeting of MUC5AC ameliorates therapeutic outcome in experimental colitis. NANOSCALE 2024; 16:5715-5728. [PMID: 38407269 DOI: 10.1039/d3nr05681c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Inflammatory bowel diseases (IBDs), which include Crohn's disease (CD) and ulcerative colitis (UC), are chronic inflammatory diseases of the gastrointestinal tract and are characterized by chronic recurrent ulceration of the bowels. Colon-targeted drug delivery systems (DDS) have received significant attention for their potential to treat IBD by improving the inflamed tissue selectivity. Herein, antiMUC5AC-decorated drug loaded nanoparticles (NP) are suggested for active epithelial targeting and selective adhesion to the inflamed tissue in experimental colitis. NPs conjugated with antiMUC5AC (anti-MUC5) were tested for their degree of bioadhesion with HT29-MTX cells by comparison with non-targeted BSA-NP conjugates. In vivo, the selectivity of bioadhesion and the influence of ligand density in bioadhesion efficiency as well as the therapeutic benefit for glucocorticoid loaded anti-MUC5-NP were studied in a murine colitis model. Quantitative adhesion analyses showed that anti-MUC5-conjugated NP exhibited a much higher binding and selectivity to inflamed tissue compared to PNA-, IgG1- and BSA-NP conjugates used as controls. This bioadhesion efficiency was found to be dependent on the ligand density, present at the NP surface. The binding specificity between anti-MUC5 ligand and inflamed tissues was confirmed by fluorescence imaging. Both anti-MUC5-NP and all other glucocorticoid containing formulations led to a significant mitigation of the experimental colitis, as became evident from the substantial reduction of myeloperoxidase activity and pro-inflammatory cytokine concentrations (TNF-α, IL-1β). Targeted NP by using anti-MUC5 appears to be a very promising tool in future treatment of various types of local disorders affecting the gastro-intestinal tract but not limited to colitis.
Collapse
Affiliation(s)
- Bernadette Riemann
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Germany
| | - Thomas Antoine
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| | - Arnaud Béduneau
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| | - Yann Pellequer
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| | - Alf Lamprecht
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Germany
| | - Brice Moulari
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| |
Collapse
|
40
|
Kong D, Qian Y, Yu B, Hu Z, Cheng C, Wang Y, Fang Z, Yu J, Xiang S, Cao L, He Y. Interaction of human dendritic cell receptor DEC205/CD205 with keratins. J Biol Chem 2024; 300:105699. [PMID: 38301891 PMCID: PMC10914487 DOI: 10.1016/j.jbc.2024.105699] [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: 09/19/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/03/2024] Open
Abstract
DEC205 (CD205) is one of the major endocytic receptors on dendritic cells and has been widely used as a receptor target in immune therapies. It has been shown that DEC205 can recognize dead cells through keratins in a pH-dependent manner. However, the mechanism underlying the interaction between DEC205 and keratins remains unclear. Here we determine the crystal structures of an N-terminal fragment of human DEC205 (CysR∼CTLD3). The structural data show that DEC205 shares similar overall features with the other mannose receptor family members such as the mannose receptor and Endo180, but the individual domains of DEC205 in the crystal structure exhibit distinct structural features that may lead to specific ligand binding properties of the molecule. Among them, CTLD3 of DEC205 adopts a unique fold of CTLD, which may correlate with the binding of keratins. Furthermore, we examine the interaction of DEC205 with keratins by mutagenesis and biochemical assays based on the structural information and identify an XGGGX motif on keratins that can be recognized by DEC205, thereby providing insights into the interaction between DEC205 and keratins. Overall, these findings not only improve the understanding of the diverse ligand specificities of the mannose receptor family members at the molecular level but may also give clues for the interactions of keratins with their binding partners in the corresponding pathways.
Collapse
Affiliation(s)
- Dandan Kong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanying Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bowen Yu
- Department of Immunology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Zhenzheng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Cheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Fang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Yu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Immune Microenvironment and Disease, Tianjin Medical University, Tianjin, China
| | - Song Xiang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Immune Microenvironment and Disease, Tianjin Medical University, Tianjin, China
| | - Longxing Cao
- School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Yongning He
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation, Shanghai, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
41
|
Swami R, Vij S, Sharma S. Unlocking the power of sugar: carbohydrate ligands as key players in nanotherapeutic-assisted targeted cancer therapy. Nanomedicine (Lond) 2024; 19:431-453. [PMID: 38288611 DOI: 10.2217/nnm-2023-0276] [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: 03/01/2024] Open
Abstract
Cancer cells need as much as 40-times more sugar than their normal cell counterparts. This sugar demand is attained by the excessive expression of inimitable transporters on the surface of cancer cells, driven by their voracious appetite for carbohydrates. Nanotechnological advances drive research utilizing ligand-directed therapeutics and diverse carbohydrate analogs. The precise delivery of these therapeutic cargos not only mitigates toxicity associated with chemotherapy but also reduces the grim toll of mortality and morbidity among patients. This in-depth review explores the potential of these ligands in advanced cancer treatment using nanoparticles. It offers a broader perspective beyond the usual ways we deliver drugs, potentially changing the way we fight cancer.
Collapse
Affiliation(s)
- Rajan Swami
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Sahil Vij
- Maharishi Markandeshwar College of Pharmacy, Maharishi Markandeshwar University, Mullana, Haryana, 133203, India
| | - Shubham Sharma
- Maharishi Markandeshwar College of Pharmacy, Maharishi Markandeshwar University, Mullana, Haryana, 133203, India
| |
Collapse
|
42
|
Wang X, Wan W, Zhang J, Lu J, Liu P. Efficient pulmonary fibrosis therapy via regulating macrophage polarization using respirable cryptotanshinone-loaded liposomal microparticles. J Control Release 2024; 366:1-17. [PMID: 38154539 DOI: 10.1016/j.jconrel.2023.12.042] [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: 10/22/2023] [Revised: 12/18/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
Lung inflammation and fibrogenesis are the two main characteristics during the development of pulmonary fibrosis (PF), which are particularly associated with pulmonary macrophages. In this context, whether cryptotanshinone (CTS) could alleviate PF through regulating macrophage polarization were preliminarily demonstrated in vitro. Then the time course of PF and its relationship with macrophage polarization was determined in BLM-induced mice based on cytokine levels in bronchoalveolar lavage fluid (BALF), lung histopathology, flow cytometric analysis, mRNA and protein expression. CTS was loaded into macrophage-targeted and responsively released mannose-modified liposomes (Man-lipo), and the liposomes were then embedded into mannitol microparticles (M-MPs) using spray drying to achieve efficient pulmonary delivery. Afterwards, how CTS regulates macrophage polarization in vivo during different time courses of PF was probed. Furthermore, the molecular mechanisms of CTS against PF by regulating macrophage polarization were elucidated in vivo and in vitro. The full-course therapy group could achieve comparable therapeutic effects compared with the positive control drug PFD group. CTS can alleviate PF through regulating macrophage polarization, mainly by inhibiting NLRP3/TGF-β1 pathway during the inflammation course and modulating MMP-9/TIMP-1 balance during the fibrosis development course, providing new insights into chronic PF treatment.
Collapse
Affiliation(s)
- Xiuhua Wang
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wei Wan
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiguo Zhang
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Jing Lu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Peiqing Liu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China.
| |
Collapse
|
43
|
Zhang W, Wan Z, Qu D, Sun W, Zhang L, Liang Y, Pan L, Jiang H, Ye Z, Wei M, Yuan L, Yang G, Jin F. Profibrogenic macrophage-targeted delivery of mitochondrial protector via exosome formula for alleviating pulmonary fibrosis. Bioact Mater 2024; 32:488-501. [PMID: 37965241 PMCID: PMC10641087 DOI: 10.1016/j.bioactmat.2023.09.019] [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: 03/17/2023] [Revised: 08/24/2023] [Accepted: 09/25/2023] [Indexed: 11/16/2023] Open
Abstract
Pulmonary fibrosis (PF) is a devastating lung disease with limited treatment options. During this pathological process, the profibrogenic macrophage subpopulation plays a crucial role, making the characterization of this subpopulation fundamentally important. The present study revealed a positive correlation between pulmonary macrophages with higher mitochondrial mass (Mømitohigh) and fibrosis. Among the Mømitohigh subpopulation of CD206+ M2, characterized by higher expression of dynamin 1-like (Drp1), as determined by flow cytometry and RNA-seq analysis, a therapeutic intervention was developed using an exosome-based formula composed of pathfinder and therapeutics. A pathfinder exosome called "exosomeMMP19 (ExoMMP19)", was constructed to display matrix metalloproteinase-19 (MMP19) on the surface to locally break down the excessive extracellular matrix (ECM) in the fibrotic lung. A therapeutic exosome called "exosome therapeutics (ExoTx)", was engineered to display D-mannose on the surface while encapsulating siDrp1 inside. Prior delivery of ExoMMP19 degraded excessive ECM and thus paved the way for ExoTx to be delivered into Mømitohigh, where ExoTx inhibited mitochondrial fission and alleviated PF. This study has not only identified Mømitohigh as profibrotic macrophages but it has also provided a potent strategy to reverse PF via a combination of formulated exosomes.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Zhuo Wan
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Di Qu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Clinical Pharmacy, General Hospital of Western Theater Command, Chengdu, Sichuan, 610083, China
| | - Wenqi Sun
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Liang Zhang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Yuan Liang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Lei Pan
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Hua Jiang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Zichen Ye
- Department of Health Service, Health Service Training Base, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Mengying Wei
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Lijun Yuan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Guodong Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Faguang Jin
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| |
Collapse
|
44
|
Ravi S, Martin LC, Krishnan M, Kumaresan M, Manikandan B, Ramar M. Interactions between macrophage membrane and lipid mediators during cardiovascular diseases with the implications of scavenger receptors. Chem Phys Lipids 2024; 258:105362. [PMID: 38006924 DOI: 10.1016/j.chemphyslip.2023.105362] [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: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The onset and progression of cardiovascular diseases with the major underlying cause being atherosclerosis, occur during chronic inflammatory persistence in the vascular system, especially within the arterial wall. Such prolonged maladaptive inflammation is driven by macrophages and their key mediators are generally attributed to a disparity in lipid metabolism. Macrophages are the primary cells of innate immunity, endowed with expansive membrane domains involved in immune responses with their signalling systems. During atherosclerosis, the membrane domains and receptors control various active organisations of macrophages. Their scavenger/endocytic receptors regulate the trafficking of intracellular and extracellular cargo. Corresponding influence on lipid metabolism is mediated by their dynamic interaction with scavenger membrane receptors and their integrated mechanisms such as pinocytosis, phagocytosis, cholesterol export/import, etc. This interaction not only results in the functional differentiation of macrophages but also modifies their structural configurations. Here, we reviewed the association of macrophage membrane biomechanics and their scavenger receptor families with lipid metabolites during the event of atherogenesis. In addition, the membrane structure of macrophages and the signalling pathways involved in endocytosis integrated with lipid metabolism are detailed. This article establishes future insights into the scavenger receptors as potential targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Kumaresan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai 600 015, India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
45
|
Tu W, Hu X, Wan R, Xiao X, Shen Y, Srikaram P, Avvaru SN, Yang F, Pi F, Zhou Y, Wan M, Gao P. Effective delivery of miR-511-3p with mannose-decorated exosomes with RNA nanoparticles confers protection against asthma. J Control Release 2024; 365:602-616. [PMID: 37996055 PMCID: PMC10872989 DOI: 10.1016/j.jconrel.2023.11.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
Our previous studies have shown that miR-511-3p treatment has a beneficial effect in alleviating allergic airway inflammation. Here, we sought to explore its therapeutic potential in animal models and gain a deeper understanding of its therapeutic value for asthma. miR-511-3p knockout mice (miR-511-3p-/-) were generated by CRISPR/Cas and showed exacerbated airway hyper-responsiveness and Th2-associated allergic airway inflammation compared with wild-type (WT) mice after exposed to cockroach allergen. RNA nanoparticles with mannose decorated EV-miR-511-3p were also created by loading miR-511-3p mimics into the mannose decorated EVs with engineered RNA nanoparticle PRNA-3WJ (Man-EV-miR-511-3p). Intra-tracheal inhalation of Man-EV-miR-511-3p, which could effectively penetrate the airway mucus barrier and deliver functional miR-511-3p to lung macrophages, successfully reversed the increased airway inflammation observed in miR-511-3p-/- mice. Through microarray analysis, complement C3 (C3) was identified as one of the major targets of miR-511-3p. C3 was increased in LPS-treated macrophages but decreased after miR-511-3p treatment. Consistent with these findings, C3 expression was elevated in the lung macrophages of an asthma mouse model but decreased in mice treated with miR-511-3p. Further experiments, including miRNA-mRNA pulldown and luciferase reporter assays, confirmed that miR-511-3p directly binds to C3 and activates the C3 gene. Thus, miR-511-3p represents a promising therapeutic target for asthma, and RNA nanotechnology reprogrammed EVs are efficient carriers for miRNA delivery for disease treatment.
Collapse
Affiliation(s)
- Wei Tu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA; Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen 518020, China; The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Xinyue Hu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA; Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Rongjun Wan
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA; Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiaojun Xiao
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yingchun Shen
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Prakhyath Srikaram
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Sai Nithin Avvaru
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Fuhan Yang
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | | | - Yufeng Zhou
- Children's Hospital and Institute of Biomedical Sciences, Fudan University, Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Mei Wan
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA.
| |
Collapse
|
46
|
Mamedov AT, Gantsova EA, Kiseleva VV, Lokhonina AV, Makarov AV, Turygina SA, Bicherova IA, Arutyunyan IV, Vishnyakova PA, Elchaninov AV, Fatkhudinov TK. Macrophage population state and proliferative activity of spleen cells under liver regeneration conditions. RUDN JOURNAL OF MEDICINE 2023; 27:441-448. [DOI: 10.22363/2313-0245-2023-27-4-441-448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Relevance. Currently, the participation of immune system cells in the regulation of reparative processes is attracting more and more attention of researchers. There is an anatomical connection between the liver and spleen by means of portal vein. Thus, cytokines and other biologically active substances can enter the liver from the spleen through the portal vein, as well as cells can migrate to the liver. However, the specific mechanisms of mutual influence of the mentioned organs, including in reparative processes, remain poorly studied. The aim of our work was to study the state of spleen monocyte-macrophage population after liver resection, as well as the proliferative activity of spleen cells during liver regeneration . Materials and Methods . The model of liver regeneration after 70 % resection in mouse was reproduced in this work. The animals were taken out of the experiment after 1, 3 and 7 days. The marker of cell proliferation Ki67 was immunohistochemically detected, the state of spleen monocyte-macrophage population was evaluated by markers CD68, CD115, CD206, F4/80 by methods of immunohistochemistry and flow cytometry. Results and Discussion . The liver regeneration had a pronounced effect on the cytoarchitectonics of the spleen. In 1 day after liver resection in the spleen there was observed a decrease in the share of Ki67+cells, according to the flow cytometry data there was a decrease in the number of CD115+cells, in 3 and 7 days there was a decrease in the number of F4/80+ macrophages. Conclusion . Liver resection causes changes in the state of cell populations of the spleen as well. First of all, to the decrease in the activity of proliferative processes in it, as well as to the changes in the state of the monocyte-macrophage system. A decrease in the content of CD115+ and F4/80+ cells in the spleen was found, which indirectly indicates the migration of monocytes/macrophages after liver resection, which can also influence the course of reparative processes in the liver.
Collapse
|
47
|
Ishibashi O, Muljo SA, Islam Z. Regulation of Macrophage Polarization in Allergy by Noncoding RNAs. Noncoding RNA 2023; 9:75. [PMID: 38133209 PMCID: PMC10745746 DOI: 10.3390/ncrna9060075] [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: 09/05/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Allergy is a type 2 immune reaction triggered by antigens known as allergens, including food and environmental substances such as peanuts, plant pollen, fungal spores, and the feces and debris of mites and insects. Macrophages are myeloid immune cells with phagocytic abilities that process exogenous and endogenous antigens. Upon activation, they can produce effector molecules such as cytokines as well as anti-inflammatory molecules. The dysregulation of macrophage function can lead to excessive type 1 inflammation as well as type 2 inflammation, which includes allergic reactions. Thus, it is important to better understand how macrophages are regulated in the pathogenesis of allergies. Emerging evidence highlights the role of noncoding RNAs (ncRNAs) in macrophage polarization, which in turn can modify the pathogenesis of various immune-mediated diseases, including allergies. This review summarizes the current knowledge regarding this topic and considers three classes of ncRNAs: microRNAs, long ncRNAs, and circular ncRNAs. Understanding the roles of these ncRNAs in macrophage polarization will provide new insights into the pathogenesis of allergies and identify potential novel therapeutic targets.
Collapse
Affiliation(s)
- Osamu Ishibashi
- Laboratory of Biological Macromolecules, Graduate School of Agriculture, Osaka Metropolitan University, Sakai 599-8531, Japan
| | - Stefan A. Muljo
- Integrative Immunobiology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Zohirul Islam
- Integrative Immunobiology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
48
|
Liu Y, Lu T, Li C, Wang X, Chen F, Yue L, Jiang C. Comparative transcriptome analysis of SARS-CoV-2, SARS-CoV, MERS-CoV, and HCoV-229E identifying potential IFN/ISGs targets for inhibiting virus replication. Front Med (Lausanne) 2023; 10:1267903. [PMID: 38143441 PMCID: PMC10739311 DOI: 10.3389/fmed.2023.1267903] [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: 07/27/2023] [Accepted: 11/13/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction Since its outbreak in December 2019, SARS-CoV-2 has spread rapidly across the world, posing significant threats and challenges to global public health. SARS-CoV-2, together with SARS-CoV and MERS-CoV, is a highly pathogenic coronavirus that contributes to fatal pneumonia. Understanding the similarities and differences at the transcriptome level between SARS-CoV-2, SARS-CoV, as well as MERS-CoV is critical for developing effective strategies against these viruses. Methods In this article, we comparatively analyzed publicly available transcriptome data of human cell lines infected with highly pathogenic SARS-CoV-2, SARS-CoV, MERS-CoV, and lowly pathogenic HCoV-229E. The host gene expression profiles during human coronavirus (HCoV) infections were generated, and the pathways and biological functions involved in immune responses, antiviral efficacy, and organ damage were intensively elucidated. Results Our results indicated that SARS-CoV-2 induced a stronger immune response versus the other two highly pathogenic HCoVs. Specifically, SARS-CoV-2 induced robust type I and type III IFN responses, marked by higher upregulation of type I and type III IFNs, as well as numerous interferon-stimulated genes (ISGs). Further Ingenuity Pathway Analysis (IPA) revealed the important role of ISGs for impeding SARS-CoV-2 infection, and the interferon/ISGs could be potential targets for therapeutic interventions. Moreover, our results uncovered that SARS-CoV-2 infection was linked to an enhanced risk of multi-organ toxicity in contrast to the other two highly pathogenic HCoVs. Discussion These findings provided valuable insights into the pathogenic mechanism of SARS-CoV-2, which showed a similar pathological feature but a lower fatality rate compared to SARS-CoV and MERS-CoV.
Collapse
Affiliation(s)
- Yuzhuang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Tianyi Lu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, University of Chinese Academy of Sciences, Beijing, China
| | - Cuidan Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Xiaotong Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Fei Chen
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, University of Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | - Liya Yue
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
49
|
Ni D, Zhou H, Wang P, Xu F, Li C. Visualizing Macrophage Phenotypes and Polarization in Diseases: From Biomarkers to Molecular Probes. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:613-638. [PMID: 38223685 PMCID: PMC10781933 DOI: 10.1007/s43657-023-00129-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 01/16/2024]
Abstract
Macrophage is a kind of immune cell and performs multiple functions including pathogen phagocytosis, antigen presentation and tissue remodeling. To fulfill their functionally distinct roles, macrophages undergo polarization towards a spectrum of phenotypes, particularly the classically activated (M1) and alternatively activated (M2) subtypes. However, the binary M1/M2 phenotype fails to capture the complexity of macrophages subpopulations in vivo. Hence, it is crucial to employ spatiotemporal imaging techniques to visualize macrophage phenotypes and polarization, enabling the monitoring of disease progression and assessment of therapeutic responses to drug candidates. This review begins by discussing the origin, function and diversity of macrophage under physiological and pathological conditions. Subsequently, we summarize the identified macrophage phenotypes and their specific biomarkers. In addition, we present the imaging probes locating the lesions by visualizing macrophages with specific phenotype in vivo. Finally, we discuss the challenges and prospects associated with monitoring immune microenvironment and disease progression through imaging of macrophage phenotypes.
Collapse
Affiliation(s)
- Dan Ni
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 201203 China
| | - Heqing Zhou
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Pengwei Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 201203 China
| | - Fulin Xu
- Minhang Hospital, Fudan University, Shanghai, 201199 China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, 201203 China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 201203 China
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Shanghai, 201203 China
| |
Collapse
|
50
|
Andriana P, Makrypidi K, Liljenbäck H, Rajander J, Saraste A, Pirmettis I, Roivainen A, Li XG. Aluminum Fluoride-18 Labeled Mannosylated Dextran: Radiosynthesis and Initial Preclinical Positron Emission Tomography Studies. Mol Imaging Biol 2023; 25:1094-1103. [PMID: 37016195 PMCID: PMC10728250 DOI: 10.1007/s11307-023-01816-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 04/06/2023]
Abstract
PURPOSE In addition to being expressed on liver sinusoidal endothelial cells, mannose receptors are also found on antigen-presenting cells, including macrophages, which are mainly involved in the inflammation process. Dextran derivatives of various sizes containing cysteine and mannose moieties have previously been labeled with 99mTc and used for single-photon emission computed tomography imaging of sentinel lymph nodes. In this study, we radiolabeled 21.3-kDa D10CM with positron-emitting 18F for initial positron emission tomography (PET) studies in rats. PROCEDURES D10CM was conjugated with 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) chelator and radiolabeled with the aluminum fluoride-18 method. The whole-body distribution kinetics and stability of the intravenously administered tracer were studied in healthy male Sprague-Dawley rats by in vivo PET/CT imaging, ex vivo gamma counting, and high-performance liquid chromatography analysis. RESULTS Al[18F]F-NOTA-D10CM was obtained with a radiochemical purity of >99% and molar activity of 9.9 GBq/μmol. At 60 minutes after injection, an average of 84% of the intact tracer was found in the blood, indicating excellent in vivo stability. The highest radioactivity concentration was seen in the liver, spleen, and bone marrow, in which mannose receptors are highly expressed under physiological conditions. The uptake specificity was confirmed with in vivo blocking experiments. CONCLUSIONS Our results imply that Al[18F]F-NOTA-D10CM is a suitable tracer for PET imaging. Further studies in disease models with mannose receptor CD206-positive macrophages are warranted to clarify the tracer's potential for imaging of inflammation.
Collapse
Affiliation(s)
- Putri Andriana
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Konstantina Makrypidi
- Institute of Nuclear and Radiological Science and Technology, Energy and Safety, NCSR "Demokritos", 15310, Athens, Greece
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, FI-20520, Turku, Finland
| | - Johan Rajander
- Accelerator Laboratory, Åbo Akademi University, FI-20520, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
- Turku PET Centre, Turku University Hospital, FI-20520, Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, FI-20520, Turku, Finland
| | - Ioannis Pirmettis
- Institute of Nuclear and Radiological Science and Technology, Energy and Safety, NCSR "Demokritos", 15310, Athens, Greece
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.
- Turku Center for Disease Modeling, University of Turku, FI-20520, Turku, Finland.
- Turku PET Centre, Turku University Hospital, FI-20520, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, FI-20520, Turku, Finland.
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, FI-20520, Turku, Finland.
- Department of Chemistry, University of Turku, FI-20014, Turku, Finland.
| |
Collapse
|