1
|
Zhao X, Qiu Y, Liang L, Fu X. Interkingdom signaling between gastrointestinal hormones and the gut microbiome. Gut Microbes 2025; 17:2456592. [PMID: 39851261 PMCID: PMC11776477 DOI: 10.1080/19490976.2025.2456592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/12/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
The interplay between the gut microbiota and gastrointestinal hormones plays a pivotal role in the health of the host and the development of diseases. As a vital component of the intestinal microecosystem, the gut microbiota influences the synthesis and release of many gastrointestinal hormones through mechanisms such as modulating the intestinal environment, producing metabolites, impacting mucosal barriers, generating immune and inflammatory responses, and releasing neurotransmitters. Conversely, gastrointestinal hormones exert feedback regulation on the gut microbiota by modulating the intestinal environment, nutrient absorption and utilization, and the bacterial biological behavior and composition. The distributions of the gut microbiota and gastrointestinal hormones are anatomically intertwined, and close interactions between the gut microbiota and gastrointestinal hormones are crucial for maintaining gastrointestinal homeostasis. Interventions leveraging the interplay between the gut microbiota and gastrointestinal hormones have been employed in the clinical management of metabolic diseases and inflammatory bowel diseases, such as bariatric surgery and fecal microbiota transplantation, offering promising targets for the treatment of dysbiosis-related diseases.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ye Qiu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Lanfan Liang
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiangsheng Fu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Mladineo I, Hrabar J. Seventy years of coexistence: Parasites and Mediterranean fish aquaculture. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110355. [PMID: 40254086 DOI: 10.1016/j.fsi.2025.110355] [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: 11/18/2024] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
What can be regarded as a seedling of the contemporary aquaculture in the Mediterranean began back in the 1950s. The development of the industry did not always align with the development of ichthyopathology, a veterinary discipline aimed at identifying and combating fish diseases. Therefore, and due to the lack of published data, we are not always able to pinpoint the first outbreaks that accompanied the increase in aquaculture production. Nonetheless, fish pathogens, and parasites in particular, have shown diversity related to host species, their farming conditions and geography. Two parasite species currently regarded as dominant in Mediterranean aquaculture are the histozoic myxozoan Enteromyxum leei and the haematophagous polyopisthocotylean Sparicotyle chrysophrii, both of which infect gilthead seabream (Sparus aurata). The interactions between parasite and host with regard to the immune activity of both have been well studied using conventional immunology and omics approaches. For the remaining parasite-fish systems, our understanding of host responses and parasite mitigation mechanisms is still vague and mostly transposed from what we know of other systems. This review compiles the knowledge on fish response to the most frequent and economically important parasites in Mediterranean aquaculture, highlights the gaps and suggests further directions.
Collapse
Affiliation(s)
- Ivona Mladineo
- Institute for Marine and Antarctic Studies, University of Tasmania, Taroona, 7053, TAS, Australia; Institute of Parasitology, Biology Centre Czech Academy of Sciences, Ceske Budejovice, 37005, Czechia.
| | - Jerko Hrabar
- Institute of Oceanography and Fisheries, Split, 21000, Croatia
| |
Collapse
|
3
|
Li Y, Chen Y, Tang Y, Yang T, Zhou P, Miao L, Chen H, Deng Y. Breaking the barriers in effective and safe Toll-like receptor stimulation via nano-immunomodulators for potent cancer immunotherapy. J Control Release 2025; 382:113667. [PMID: 40157608 DOI: 10.1016/j.jconrel.2025.113667] [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: 12/02/2024] [Revised: 02/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy is an emerging strategy that awakens the intrinsic immune system for cancer treatment. Generally, successful immunotherapy of malignant tumours relies on the effective production of tumour-associated antigens and their lymph node delivery, antigen processing and presentation for T-cell activation, and the dismantling of the immunosuppressive tumour microenvironment. Toll-like receptor (TLR) agonists are potent stimulants in cancer immunotherapy, which can directly activate antigen-presenting cells (APCs) and further induce T cell activation for antitumour immune response and convert immunosuppressive tumour microenvironment to an immunogenic one for cooperative tumour ablation. However, TLR agonists for effective cancer immunotherapy have encountered essential challenges, such as insufficient immune activation and systemic side effects. In recent years, nano-immunomodulators with TLR agonists have been employed for tumour- and/or lymph node-targeted immune activation to improve the antitumour immune response and alleviate their systemic toxicities, providing a promising strategy for enhanced cancer immunotherapy. Herein, we introduce the recent progress in developing various TLR nano-immunomodulators for cancer immunotherapy via APC activation and tumour microenvironment remodelling. Upon elucidating the rational design principles of nano-immunomodulators, we elucidate the advancement of TLR nanoagonists to break the barriers in effective and safe Toll-like receptor stimulation for potent cancer immunotherapy.
Collapse
Affiliation(s)
- Yaoqi Li
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yitian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yong'an Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tao Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ping Zhou
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou 215006, China.
| | - Huabing Chen
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China.
| |
Collapse
|
4
|
Peng Y, Lin C, Zhang B, Yan L, Zhang B, Zhao C, Qiu L. Characteristics and preliminary immune function of SRA5 in Lateolabrax maculatus. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110266. [PMID: 40064212 DOI: 10.1016/j.fsi.2025.110266] [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: 01/01/2025] [Revised: 02/26/2025] [Accepted: 03/07/2025] [Indexed: 03/26/2025]
Abstract
Scavenger receptors (SRs) are crucial for pattern recognition in the innate immune system. However, the role of Scavenger Receptors class A member 5 (SRA5) in the immunological response of bony fish to pathogen invasion remains unclear. This study identified and characterized the SRA5 of Lateolabrax maculatus (LmSRA5) from its transcriptome database. LmSRA5 has a 1494 bp open reading frame, encodes 497 amino acids, has a molecular weight of 55.01 kDa, and contains a collagen domain and a conserved Scavenger Receptor Cysteine-Rich domain. LmSRA5 exhibited high sequence similarity to previously reported SRA5 genes. LmSRA5 exhibited high sequence similarity to previously reported SRA5 genes. LmSRA5 is primarily localized in the cytoplasm, with its encoded proteins distributed in both the cytoplasm and the cell membrane. LmSRA5 was expressed in all tissues. The highest expression was observed in the pituitary gland, with significant levels in the stomach, intestines, liver, and kidney. LmSRA5 expression in the head kidney, spleen, blood, and intestines initially increased, then decreased following infection with Aeromonas veronii. The binding affinity of LmSRA5 for A. veronii was enhanced by increasing concentrations of the extracellular domain recombinant LmSRA5. Knockdown and overexpression experiments in liver cells demonstrated that LmSRA5 significantly regulates the expression of IL-8 and c-Jun. LmSRA5 participates in the immune response by recognizing pathogen-associated molecular patterns (PAMPs) and contributes to immune regulation through modulation IL-8 and c-Jun. This study offers valuable insights into the role of SRA5 in pathogen resistance and immune regulation in bony fish, thereby contributing to the advancement of aquaculture under escalating disease pressures.
Collapse
Affiliation(s)
- Yangtao Peng
- College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China
| | - Changhong Lin
- College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China.
| | - Lulu Yan
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China.
| | - Chao Zhao
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China
| | - Lihua Qiu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China; Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, Chinese Academy of Fishery Science, Beijing, PR China.
| |
Collapse
|
5
|
Shome A, Ali S, Roy D, Dey S, Sinha S, Barman P, Kumar A, Chakroborty R, Haydar MS, Roy S, Ghosh S, Roy MN. Phyto-assisted eco-benevolent synthesis of oxidase-mimic Cu-Mn 3O 4 as an antibacterial and antiproliferative agent. Bioprocess Biosyst Eng 2025; 48:899-912. [PMID: 40180623 DOI: 10.1007/s00449-025-03149-x] [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: 09/18/2024] [Accepted: 03/09/2025] [Indexed: 04/05/2025]
Abstract
In recent years, the marked augment of antibiotic resistance hampered the development of antibacterial agent. Nanozymes by their in situ ROS production capability oxidize cellular substances of bacterial cell and eliminate MDR bacteria. Therefore, synthesis of effective nanozymes from green precursors is rarely reported, so the prime objective of this study was to synthesize Cu-Mn3O4 nanozymes from aqueous extracts of medicinal plant Azadirachta indica via co-precipitation approach and to endorse their biomedical applications. The synthesized materials were characterized by X-ray diffraction (XRD), Fourier Transform Infrared spectrometer (FTIR), Scanning Electron Images (SEM), and Field-Emission Scanning Electron Microscopy (FESEM) images. X-ray Diffraction (XRD) patterns revealed the formation of hausmannite Mn3O4 crystal system. Fourier Transform Infrared spectrometer (FTIR) spectra revealed functional groups on the surface nanoparticles for their stabilization. Energy-Dispersive X-ray spectroscopy (EDAX) profile confirmed the existence of desired elements in the synthesized nanozymes. B1 mimics oxidase enzyme most effectively with Km = 0.175 mM and Vmax = 10.34 µM/min. The low Km and high Vmax indicates the strong binding affinity and high catalytic activity. From the agar diffusion antibacterial assay, it can be concluded that B3 is the most potent antibacterial agent specifically against Gram-positive bacteria Bacillus subtilis with inhibition zone of 27 mm at 250 µg/mL. Their cytotoxic activities on neuroblastoma (SHSY5) cell line were investigated for the first time. The data revealed that synthesized nanooctahedrons possess a significant cytotoxicity against cancer cell lines SHSY5Y (IC50 = 137.47 ± 14.11 µg/mL) and SKOV3 (IC50 = 72.72 ± 9.33 µg/mL). Overall, with increasing Cu amount, the percentage growth inhibition of Mn3O4 crystal system enhanced. The improved antibacterial activity and cytotoxicity is due to synergy between metal and phytochemicals. Radical scavenging activity of synthesized nanozymes is comparatively lower than their green source and the comparatively lower IC50 values of B1, 234.12 ± 15.13 and 220.12 ± 10.37 respectively, which indicates that it is more active in scavenging DPPH and ABTS radical. B2 (IC50 = 310.56 ± 5.92 µg/mL) and B3 (IC50 = 43.56 ± 3.03 µg/mL) scavenge superoxide radicals and FRAP more effectively. It is noticed that synthesized nanozymes have greater antibacterial and anticancer activity but low scavenging ability compared to green extract. Thus, Cu-Mn3O4 NPs from Azadirachta indica leaf extract could be utilized as a replacement of potential antibiotic drug candidate against MDR bacteria and in cancer avenues.
Collapse
Affiliation(s)
- Ankita Shome
- Department of Chemistry, University of North Bengal, Darjeeling, India
| | - Salim Ali
- Department of Chemistry, University of North Bengal, Darjeeling, India
| | - Debadrita Roy
- Department of Chemistry, University of North Bengal, Darjeeling, India
| | - Sangita Dey
- Department of Biotechnology, University of North Bengal, Darjeeling, 734013, India
| | - Shilpa Sinha
- Department of Biotechnology, University of North Bengal, Darjeeling, 734013, India
| | - Partha Barman
- Department of Biotechnology, University of North Bengal, Darjeeling, 734013, India
| | - Anoop Kumar
- Department of Biotechnology, University of North Bengal, Darjeeling, 734013, India
| | - Ranadhir Chakroborty
- Department of Biotechnology, University of North Bengal, Darjeeling, 734013, India
| | - Md Salman Haydar
- Department of Botany, University of North Bengal, Siliguri, West Bengal, India
| | - Swarnendu Roy
- Department of Botany, University of North Bengal, Siliguri, West Bengal, India
| | - Shibaji Ghosh
- CSIR-Central Salt and Marine Chemicals Research Institute, G. B Marg, Bhavnagar, Gujarat, 364002, India
| | - Mahendra Nath Roy
- Department of Chemistry, University of North Bengal, Darjeeling, India.
| |
Collapse
|
6
|
Parolini C. Sepsis and high-density lipoproteins: Pathophysiology and potential new therapeutic targets. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167761. [PMID: 40044061 DOI: 10.1016/j.bbadis.2025.167761] [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: 10/14/2024] [Revised: 01/19/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
In 2020, sepsis has been defined a worldwide health major issue (World Health Organization). Lung, urinary tract and abdominal cavity are the preferred sites of sepsis-linked infection. Research has highlighted that the advancement of sepsis is not only related to the presence of inflammation or microbial or host pattern recognition. Clinicians and researchers now recognized that a severe immunosuppression is also a common feature found in patients with sepsis, increasing the susceptibility to secondary infections. Lipopolysaccharides (LPS) are expressed on the cell surface of Gram-negative, whereas Gram-positive bacteria express peptidoglycan (PGN) and lipoteichoic acid (LTA). The main mechanism by which LPS trigger host innate immune responses is binding to TLR4-MD2 (toll-like receptor4-myeloid differentiation factor 2), whereas, PGN and LTA are exogenous ligands of TLR2. Nucleotide-binding oligomerization domain (NOD)-like receptors are the most well-characterized cytosolic pattern recognition receptors, which bind microbial molecules, endogenous by-products and environmental triggers. It has been demonstrated that high-density lipoproteins (HDL), besides their major role in promoting cholesterol efflux, possess diverse pleiotropic properties, ranging from a modulation of the immune system to anti-inflammatory, anti-apoptotic, and anti-oxidant functions. In addition, HDL are able at i) binding LPS, preventing the activating of TLR4, and ii) inducing the expression of ATF3 (Activating transcription factor 3), a negative regulator of the TLR signalling pathways, contributing at justifying their capacity to hamper infection-based illnesses. Therefore, reconstituted HDL (rHDL), constituted by apolipoprotein A-I/apolipoprotein A-IMilano complexed with phospholipids, may be considered as a new therapeutic tool for the management of sepsis.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", via Balzaretti 9 - Università degli Studi di Milano, 20133 Milano, Italy.
| |
Collapse
|
7
|
Lu Z, Jin M, Chen S, Wang X, Sun F, Zhang Q, Zhao Z, Wu J, Yang J, Dai Q. Physics-driven self-supervised learning for fast high-resolution robust 3D reconstruction of light-field microscopy. Nat Methods 2025:10.1038/s41592-025-02698-z. [PMID: 40355725 DOI: 10.1038/s41592-025-02698-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Light-field microscopy (LFM) and its variants have significantly advanced intravital high-speed 3D imaging. However, their practical applications remain limited due to trade-offs among processing speed, fidelity, and generalization in existing reconstruction methods. Here we propose a physics-driven self-supervised reconstruction network (SeReNet) for unscanned LFM and scanning LFM (sLFM) to achieve near-diffraction-limited resolution at millisecond-level processing speed. SeReNet leverages 4D information priors to not only achieve better generalization than existing deep-learning methods, especially under challenging conditions such as strong noise, optical aberration, and sample motion, but also improve processing speed by 700 times over iterative tomography. Axial performance can be further enhanced via fine-tuning as an optional add-on with compromised generalization. We demonstrate these advantages by imaging living cells, zebrafish embryos and larvae, Caenorhabditis elegans, and mice. Equipped with SeReNet, sLFM now enables continuous day-long high-speed 3D subcellular imaging with over 300,000 volumes of large-scale intercellular dynamics, such as immune responses and neural activities, leading to widespread practical biological applications.
Collapse
Affiliation(s)
- Zhi Lu
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
- Beijing Key Laboratory of Cognitive Intelligence, Tsinghua University, Beijing, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
- Zhejiang Hehu Technology, Hangzhou, China
- Hangzhou Zhuoxi Institute of Brain and Intelligence, Hangzhou, China
| | - Manchang Jin
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
- Zhejiang Hehu Technology, Hangzhou, China
- Hangzhou Zhuoxi Institute of Brain and Intelligence, Hangzhou, China
- School of Information Science and Technology, Fudan University, Shanghai, China
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
- Shanghai Innovation Institute, Shanghai, China
| | - Shuai Chen
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoge Wang
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Feihao Sun
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Qi Zhang
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Zhifeng Zhao
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Jiamin Wu
- Department of Automation, Tsinghua University, Beijing, China.
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China.
- Beijing Key Laboratory of Cognitive Intelligence, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- Beijing Visual Science and Translational Eye Research Institute (BERI), Beijing, China.
- Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing, China.
| | - Jingyu Yang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China.
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing, China.
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China.
- Beijing Key Laboratory of Cognitive Intelligence, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing, China.
| |
Collapse
|
8
|
Rahmani NR, Jahanmard F, Hassani Najafabadi A, Flapper J, Dogan O, Khodaei A, Storm G, Croes M, Kruyt MC, Gawlitta D, Weinans H, Mastrobattista E, Amin Yavari S. Local delivery of lipid-based nanoparticles containing microbial nucleic acid for osteoimmunomodulation. Eur J Pharm Sci 2025; 208:107050. [PMID: 39988262 DOI: 10.1016/j.ejps.2025.107050] [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: 10/24/2024] [Revised: 01/27/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
Osteoimmunomodulation is a strategy to promote bone regeneration in implants by modifying the immune environment. CpG-containing oligonucleotides type C (CpG ODN C) and Polyinosinic:polycytidylic acid (Poly[I:C]) are analogs of microbial nucleic acids that have been studied for various immunotherapeutic applications. This research investigates the potential of CpG ODN C and Poly(I:C) as an osteoimmunomodulatory agent for bone regenerative purposes. We encapsulated each nucleic acid in a lipid-based nanoparticle to facilitate the delivery into intracellular pathogen recognition receptors in immune cells. The lipid-based nanoparticles were ±250 nm in size with a negative charge (-36 to -40 mV) and an encapsulation efficiency of ±60 %. Lipid-based nanoparticles containing nucleic acids, Lip/CpG ODN C and Lip/Poly(I:C), increased the production of TNF, IL-6, and IL-10 by primary human macrophages compared to free-form nucleic acids. Conditioned medium from macrophages treated with CpG ODN C (10 µg/ml) and Lip/CpG ODN C (0.1, 1, and 10 µg/ml) promoted osteoblast differentiation of human mesenchymal stromal cells by 2.6-fold and 3-fold, respectively; no effect was seen for Lip/Poly(I:C). Bone implants were prepared, consisting of a biphasic calcium phosphate scaffold, bone morphogenetic protein (BMP) 2, and lipid-based nanoparticles suspended in gelatin methacryloyl (GelMA) hydrogel. Implants were evaluated for de novo bone formation in an extra-skeletal implantation model in rabbits for 5 weeks. Based on the particles suspended in GelMA, six groups of implants were prepared: Lip/CpG ODN C, Lip/Poly(I:C), Lip (empty), CpG ODN C, Poly(I:C), and a control group consisting of empty GelMA. After 5 weeks, healthy bone tissue formed in all of the implants with active osteoblast and osteoclast activity, however, the amount of new bone volume and scaffold degradation were similar for all implants. We suggest that the working concentrations of the nucleic acids employed were inadequate to induce a relevant inflammatory response. Additionally, the dosage of BMP-2 used may potentially mask the immune-stimulatory effect. Lip/CpG ODN C holds potential as a bioactive agent for osteoimmunomodulation, although further in vivo demonstration should corroborate the current in vitro findings.
Collapse
Affiliation(s)
- N R Rahmani
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, University Utrecht, Heidelberglaan 8, CS 3584, Utrecht, the Netherlands.
| | - F Jahanmard
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - A Hassani Najafabadi
- Terasaki Institute for Biomedical Innovation, 21100 Erwin St., Woodland Hills, 91367, Los Angeles, United States.
| | - J Flapper
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - O Dogan
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - A Khodaei
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, University Utrecht, Heidelberglaan 8, CS 3584, Utrecht, the Netherlands.
| | - G Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - M Croes
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands.
| | - M C Kruyt
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Department of Developmental Biomedical Engineering, Twente University, Drienerlolaan 5, NB 7522, Enschede, the Netherlands.
| | - D Gawlitta
- Regenerative Medicine Center Utrecht, University Utrecht, Heidelberglaan 8, CS 3584, Utrecht, the Netherlands; Department of Oral and Maxillofacial Surgery, Prosthodontics and Special Dental Care, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands.
| | - H Weinans
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Department of Biomechanical Engineering, Technical University Delft, Mekelweg 2, CD 2628, Delft, the Netherlands.
| | - E Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, CG 3584, Utrecht, the Netherlands.
| | - S Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, GA 3508, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, University Utrecht, Heidelberglaan 8, CS 3584, Utrecht, the Netherlands; Terasaki Institute for Biomedical Innovation, 21100 Erwin St., Woodland Hills, 91367, Los Angeles, United States.
| |
Collapse
|
9
|
Song M, Zhang S, Gan Y, Ding T, Li Z, Fan X. Poria cocos Polysaccharide Reshapes Gut Microbiota to Regulate Short-Chain Fatty Acids and Alleviate Neuroinflammation-Related Cognitive Impairment in Alzheimer's Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:10316-10330. [PMID: 40254847 DOI: 10.1021/acs.jafc.5c01042] [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: 04/22/2025]
Abstract
Evidence indicates that Poria cocos polysaccharide (PCP) improves cognitive impairment in Alzheimer's disease (AD); however, its underlying mechanism, particularly its relationship with the gut microbiota, remains unclear. In the current study, we aimed to investigate the mechanism of PCP in improving cognitive impairment in AD. The results demonstrated that PCP markedly enhanced cognitive function and mitigated AD-related pathological alterations in 3 × Tg-AD mice. PCP treatment reversed the age-dependent gut microbiota dysbiosis in 3 × Tg-AD mice by 16S rDNA sequencing. The contents of propanoic acid, butanoic acid and isohexanoic acid were increased by short-chain fatty acid determination. In addition, PCP could restore both the intestinal barrier and the blood-brain barrier, as demonstrated by immunofluorescence staining of tight junction proteins. Furthermore, PCP alleviated systemic inflammation and neuroinflammation, as evidenced by reduced LPS levels in circulation and decreased IL-6 levels in the brain, likely by inhibiting the TLR4/NF-κB signaling pathway. In conclusion, PCP can reshape gut microbiota to regulate short-chain fatty acids and alleviate neuroinflammation-related cognitive impairment in AD mice.
Collapse
Affiliation(s)
- Meiying Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yuxin Gan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Tao Ding
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhu Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
10
|
Nazir A, Hussain FHN, Nadeem Hussain TH, Al Dweik R, Raza A. Therapeutic targeting of the host-microbiota-immune axis: implications for precision health. Front Immunol 2025; 16:1570233. [PMID: 40364844 PMCID: PMC12069365 DOI: 10.3389/fimmu.2025.1570233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/24/2025] [Indexed: 05/15/2025] Open
Abstract
The human body functions as a complex ecosystem, hosting trillions of microbes that collectively form the microbiome, pivotal in immune system regulation. The host-microbe immunological axis maintains homeostasis and influences key physiological processes, including metabolism, epithelial integrity, and neural function. Recent advancements in microbiome-based therapeutics, including probiotics, prebiotics and fecal microbiota transplantation, offer promising strategies for immune modulation. Microbial therapies leveraging microbial metabolites and engineered bacterial consortia are emerging as novel therapeutic strategies. However, significant challenges remain, including individual microbiome variability, the complexity of host-microbe interactions, and the need for precise mechanistic insights. This review comprehensively examines the host microbiota immunological interactions, elucidating its mechanisms, therapeutic potential, and the future directions of microbiome-based immunomodulation in human health. It will also critically evaluate challenges, limitations, and future directions for microbiome-based precision medicine.
Collapse
Affiliation(s)
- Asiya Nazir
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | | | | | - Rania Al Dweik
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Afsheen Raza
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
11
|
Wu S, Xu W, Shan X, Sun L, Liu S, Sun X, Li S, Hou X, Bo X, Peng C, Huang B, Xu H, Yue W. Targeting Splenic Myeloid Cells with Nanobiologics to Prevent Postablative Pancreatic Cancer Recurrence via Inducing Antitumor Peripheral Trained Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2413562. [PMID: 40289661 DOI: 10.1002/advs.202413562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/27/2025] [Indexed: 04/30/2025]
Abstract
Minimally invasive irreversible electroporation ablation shows promise for pancreatic cancer (PCa), but the high recurrence and metastasis rates pose a therapeutic challenge for loco-regional ablation treatment. Immunotherapy holds promise for preventing tumor recurrence, however, its efficacy against PCa remains limited. Here, using a preclinical model of PCa, it is identified that tumor development dramatically restructures the splenic immune landscape characterized by increased frequency of myeloid cells. Further, nanobiologics with high affinity for splenic myeloid cells using erythrocyte membrane fused with apoA1-modified liposomes are presented. Biocompatible CaCO3 nanoparticles are incorporated to serve as a release reservoir of immunomodulatory therapeutics (muramyl dipeptide, MDP). The nanobiologics, MDCa@RBC-Alipo, induce antitumor-trained immunity by epigenetically and metabolically rewiring splenic myeloid cells, thereby overcoming the immunosuppressive tumor microenvironment in residual PCa following irreversible electroporation ablation. This approach enhances the therapeutic efficacy of aPD-L1 and significantly inhibits tumor recurrence and hemorrhagic ascites development. The trafficking of MDP directly to the spleen highlights a previously uncharacterized pathway for inducing peripheral trained immunity, thereby presenting a novel therapeutic approach for locally advanced PCa treatment.
Collapse
Affiliation(s)
- Shengbo Wu
- Ultrasound Department, Zhejiang Hospital, No. 1229 Gudun Road, Xihu District, Hangzhou, Zhejiang Province, 310013, P. R. China
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Weichen Xu
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Xuexia Shan
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Liping Sun
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Shuo Liu
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Xixi Sun
- Ultrasound Department, Zhejiang Hospital, No. 1229 Gudun Road, Xihu District, Hangzhou, Zhejiang Province, 310013, P. R. China
| | - Shaoyue Li
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Xiaodong Hou
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Xiaowan Bo
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Chengzhong Peng
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| | - Bin Huang
- Ultrasound Department, Zhejiang Hospital, No. 1229 Gudun Road, Xihu District, Hangzhou, Zhejiang Province, 310013, P. R. China
| | - Huixiong Xu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, P. R. China
| | - Wenwen Yue
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Shanghai, 200072, P. R. China
| |
Collapse
|
12
|
Lee Y, Ishikawa T, Lee H, Lee B, Ryu C, Davila Mejia I, Kim M, Lu G, Hong Y, Feng M, Shin H, Meloche S, Locksley RM, Koltsova E, Grivennikov SI, Heiman M, Choi GB, Huh JR. Brain-wide mapping of immune receptors uncovers a neuromodulatory role of IL-17E and the receptor IL-17RB. Cell 2025; 188:2203-2217.e17. [PMID: 40199322 PMCID: PMC12063771 DOI: 10.1016/j.cell.2025.03.006] [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: 03/04/2024] [Revised: 11/17/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025]
Abstract
Cytokines interact with their receptor complexes to orchestrate diverse processes-from immune responses to behavioral modulation. Interleukin-17A (IL-17A) mediates protective immune responses by binding to IL-17 receptor A (IL-17RA) and IL-17RC subunits. IL-17A also modulates social interaction, yet the role of cytokine receptors in this process and their expression in the brain remains poorly characterized. Here, we mapped the brain-region-specific expression of all major IL-17R subunits and found that in addition to IL-17RA, IL-17RB-but not IL-17RC-plays a role in social behaviors through its expression in the cortex. We further showed that IL-17E, expressed in cortical neurons, enhances social interaction by acting on IL-17RA- and IL-17RB-expressing neurons. These findings highlight an IL-17 circuit within the cortex that modulates social behaviors. Thus, characterizing spatially restricted cytokine receptor expression can be leveraged to elucidate how cytokines function as critical messengers mediating neuroimmune interactions to shape animal behaviors.
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Tomoe Ishikawa
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hyeseung Lee
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Byeongjun Lee
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Changhyeon Ryu
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Irene Davila Mejia
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Minjin Kim
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Guangqing Lu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yujin Hong
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Mengyang Feng
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hyeyoon Shin
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer (IRIC), Montreal, QC, Canada
| | - Richard M Locksley
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Ekaterina Koltsova
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sergei I Grivennikov
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Myriam Heiman
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gloria B Choi
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Human Biology Microbiome Quantum Research Center (Bio2Q), Keio University, Tokyo, Japan; Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA.
| |
Collapse
|
13
|
Li L, Liu J, Lu J, Wu J, Zhang X, Ma T, Wu X, Zhu Q, Chen Z, Tai Z. Interventions in cytokine signaling: novel horizons for psoriasis treatment. Front Immunol 2025; 16:1573905. [PMID: 40303401 PMCID: PMC12037536 DOI: 10.3389/fimmu.2025.1573905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Intricate interactions between immune cells and cytokines define psoriasis, a chronic inflammatory skin condition that is immunological-mediated. Cytokines, including interleukins (ILs), interferons (IFNs), tumor necrosis factors (TNFs), chemokines, and transforming growth factor-β (TGF-β), are essential for controlling cellular activity and immunological responses, maintaining homeostasis and contributing to the pathogenesis of psoriasis. These molecules modulate the immune microenvironment by either promoting or suppressing inflammation, which significantly impacts therapeutic outcomes. Recent research indicates that treatment strategies targeting cytokines and chemokines have significant potential, offering new approaches for regulating the immune system, inhibiting the progression of psoriasis, and reducing adverse effects of traditional therapies. This review consolidates current knowledge on cytokine and chemokine signaling pathways in psoriasis and examines their significance in treatment. Specific attention is given to cytokines like IL-17, IL-23, and TNF-α, underscoring the necessity for innovative therapies to modulate these pathways and address inflammatory processes. This review emphasizes the principal part of cytokines in the -pathological process of psoriasis and explores the challenges and opportunities they present for therapeutic intervention. Furthermore, we examine recent advancements in targeted therapies, with a particular focus on monoclonal antibodies, in ongoing research and clinical trials.
Collapse
Affiliation(s)
- Lisha Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Jiaye Lu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Junchao Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Tianyou Ma
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Xiying Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Dalal N, Dhandapani H, Ingle A, Sharma D, Tayalia P. Functionalized Poly(ethylene Glycol) Diacrylate Scaffolds for In Situ Immunomodulation of Dendritic Cells Targeting Melanoma Tumor. ACS Biomater Sci Eng 2025; 11:2396-2407. [PMID: 40048381 DOI: 10.1021/acsbiomaterials.4c02036] [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: 04/15/2025]
Abstract
Various immunotherapeutic strategies are being developed to fight cancer, which is one of the leading causes of mortality. Dendritic cells (DCs), being professional antigen-presenting cells, after efficient manipulation with tumor-associated antigens, can lead to effective T-cell recruitment and activation at the tumor site, resulting in cytotoxic T-cell-mediated cancer cell killing. To circumvent the inefficiencies of ex vivo DC modification and patient infusion, an alternative strategy involving in situ DC activation has been explored here. Here, the vaccine components are tumor lysates, as antigens, and polyinosinic:polycytidylic acid (poly(I:C)), a toll-like receptor-3 (TLR3) agonist, as an adjuvant. Our in vitro studies demonstrate that complexing poly(I:C) with a carrier molecule, chitosan, enhances its stability and accessibility to TLR3 in the DC endosomal membrane. Material-based localized delivery of immunomodulatory factors is known to improve their stability and reduce their off-target side effects. Here, PEGDA-PLL-based macroporous scaffolds allow easy recruitment of host cells, thereby enabling effective interaction between the vaccine components loaded on them and the infiltrating immune cells. The vaccine components present in the scaffold facilitate efficient DC activation and migration, leading to subsequent T-cell activation and antitumor response, as shown by our in vivo studies.
Collapse
Affiliation(s)
- Neha Dalal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Hemavathi Dhandapani
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Arvind Ingle
- Tata Memorial Centre Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Deepak Sharma
- Radiation Biology and Health Science Division, Bhabha Atomic Research Center, Mumbai 400085, India
| | - Prakriti Tayalia
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
15
|
Chai C, Sultan E, Sarkar SR, Zhong L, Sarfati DN, Gershoni-Yahalom O, Jacobs-Wagner C, Rosental B, Wang B. Explosive cytotoxicity of 'ruptoblasts' bridges hormonal surveillance and immune defense. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645876. [PMID: 40236000 PMCID: PMC11996342 DOI: 10.1101/2025.03.28.645876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cytotoxic killing is an essential immune function, yet its cellular mechanisms have been characterized in only a few model species. Here, we show that planarian flatworms harness a unique cytotoxic strategy. In planarians, activin, a hormone regulating regeneration and reproduction, also acts as an inflammatory cytokine. Overactivation of activin signaling - through protein injection, genetic chimerism, or bacterial infection - triggers 'ruptoblasts', an undocumented immune cell type, to undergo 'ruptosis', a unique mode of cell bursting that eliminates nearby cells and bacteria in mere minutes, representing one of the fastest cytotoxic mechanisms observed. Ablating ruptoblasts suppresses inflammation but compromises bacterial clearance, highlighting ruptoblasts' broad-spectrum immune functions. We further identified ruptoblast-like cells in diverse basal bilaterians, unveiling an alternative strategy that couples hormonal regulation with immune defense and expanding the landscape of evolutionary immune innovations.
Collapse
|
16
|
Fu Z, Lin S, Chen H, Guo H, Li J, Chen Y, Lu Y, Liu J, Huang W, Pang Y. Generating Self-Adjuvated Nanofiber Vaccines by Coating Bacterial Flagella with Antigens. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415887. [PMID: 39981905 DOI: 10.1002/adma.202415887] [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: 10/17/2024] [Revised: 01/28/2025] [Indexed: 02/22/2025]
Abstract
Bacteria-based vaccines have received increasing attention given the ability to induce strong systemic immune responses. However, the application of bacteria as therapeutic agents inevitably suffers from infection-associated side effects due to the living characteristics. Here, the use of bacteria-derived flagella is described to construct self-adjuvated nanofiber vaccines. With the help of charge-reversal mediated by decoration with cationic polymers, the flagella can be coated with negatively charged antigens through electrostatic interaction. By virtue of the large aspect ratio, the resulting nanofiber vaccines show prolonged retention at the injection site and increased uptake by dendritic cells and macrophages. Thanks to the innate immunogenicity, self-adjuvated flagella robustly promote dendritic cell maturation and macrophage polarization, resulting in the elicitation of antigen-specific T-cell and B-cell immune responses. In ovalbumin-overexpressing melanoma-bearing mice, immunization with ovalbumin-carried vaccines not only exhibits a favorable tolerance, but also displays superior inhibition efficacies on tumor growth and metastasis separately under the therapeutic and prophylactic settings. The flexibility of this approach is further demonstrated for vaccine fabrication by coating with the SARS-CoV-2 Spike protein S1 subunit. Bacterial flagella-based self-adjuvated nanofiber platform proposes a versatile strategy to develop various vaccines for disease prevention and treatment.
Collapse
Affiliation(s)
- Zhenzhen Fu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huan Chen
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haiyan Guo
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Juanjuan Li
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanmei Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yue Lu
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Xiangfu Laboratory, Jiaxing, Zhejiang, 314102, China
| | - Yan Pang
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
17
|
Zhang K, Jagannath C. Crosstalk between metabolism and epigenetics during macrophage polarization. Epigenetics Chromatin 2025; 18:16. [PMID: 40156046 PMCID: PMC11954343 DOI: 10.1186/s13072-025-00575-9] [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: 11/24/2024] [Accepted: 02/17/2025] [Indexed: 04/01/2025] Open
Abstract
Macrophage polarization is a dynamic process driven by a complex interplay of cytokine signaling, metabolism, and epigenetic modifications mediated by pathogens. Upon encountering specific environmental cues, monocytes differentiate into macrophages, adopting either a pro-inflammatory (M1) or anti-inflammatory (M2) phenotype, depending on the cytokines present. M1 macrophages are induced by interferon-gamma (IFN-γ) and are characterized by their reliance on glycolysis and their role in host defense. In contrast, M2 macrophages, stimulated by interleukin-4 (IL-4) and interleukin-13 (IL-13), favor oxidative phosphorylation and participate in tissue repair and anti-inflammatory responses. Metabolism is tightly linked to epigenetic regulation, because key metabolic intermediates such as acetyl-coenzyme A (CoA), α-ketoglutarate (α-KG), S-adenosylmethionine (SAM), and nicotinamide adenine dinucleotide (NAD+) serve as cofactors for chromatin-modifying enzymes, which in turn, directly influences histone acetylation, methylation, RNA/DNA methylation, and protein arginine methylation. These epigenetic modifications control gene expression by regulating chromatin accessibility, thereby modulating macrophage function and polarization. Histone acetylation generally promotes a more open chromatin structure conducive to gene activation, while histone methylation can either activate or repress gene expression depending on the specific residue and its methylation state. Crosstalk between histone modifications, such as acetylation and methylation, further fine-tunes macrophage phenotypes by regulating transcriptional networks in response to metabolic cues. While arginine methylation primarily functions in epigenetics by regulating gene expression through protein modifications, the degradation of methylated proteins releases arginine derivatives like asymmetric dimethylarginine (ADMA), which contribute directly to arginine metabolism-a key factor in macrophage polarization. This review explores the intricate relationships between metabolism and epigenetic regulation during macrophage polarization. A better understanding of this crosstalk will likely generate novel therapeutic insights for manipulating macrophage phenotypes during infections like tuberculosis and inflammatory diseases such as diabetes.
Collapse
Affiliation(s)
- Kangling Zhang
- Department of Pharmacology and Toxicology, School of Medicine, University of Texas Medical Branch, Galveston, TX, USA.
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA.
| |
Collapse
|
18
|
Saha S, Sachivkina N, Kuznetsova O, Neborak E, Zhabo N. Elucidating the Role of Nrf2 Signaling Pathway in Mycoplasma Infections. FRONT BIOSCI-LANDMRK 2025; 30:28286. [PMID: 40302330 DOI: 10.31083/fbl28286] [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: 11/18/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 05/02/2025]
Abstract
Mycoplasmas are the smallest cell-wall-less self-replicating prokaryotes. Mycoplasma species can be found within and outside cells as "silent parasites" that live intracellularly and as membrane surface parasites. The pathogen's impact on respiratory health seems primarily caused by its capacity to alter immune responses, cause airway inflammation, and damage epithelial barriers. Much progress has been made in understanding Mycoplasma-induced inflammation and oxidative stress. However, there are still issues in therapeutic management, such as the development of strains that are resistant to antibiotics, the shortcomings of the available diagnostic techniques, and possible long-term respiratory consequences. On the other hand, to combat oxidative stress, inflammation, and metabolic abnormalities, activation of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is becoming a more appealing therapeutic strategy. Nrf2 activation coordinates a thorough defense through its transcriptional targets, enabling adaptability and survival under a variety of cellular stressors. Nrf2 is regarded as a therapeutic target, and pharmacological Nrf2 activators have demonstrated protective effects in multiple pathological consequences and advantages in clinical trials. In this review, we discussed the rationale for targeting Nrf2 in a series of inflammatory responses caused by Mycoplasma species.
Collapse
Affiliation(s)
- Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, 281406 Mathura, Uttar Pradesh, India
| | - Nadezhda Sachivkina
- Department of Microbiology V.S. Kiktenko, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Olga Kuznetsova
- Department of Biochemistry T.T. Berezov, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Neborak
- Department of Biochemistry T.T. Berezov, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Natallia Zhabo
- Department of Foreign Languages, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
19
|
Raza ML. The stress-immune system axis: Exploring the interplay between stress and immunity. PROGRESS IN BRAIN RESEARCH 2025; 291:289-317. [PMID: 40222784 DOI: 10.1016/bs.pbr.2025.01.003] [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: 04/15/2025]
Abstract
The chapter talks about how our body and mind respond to stress and how it affects our immune system. Stress reactions, especially the fight-or-flight reaction, are helpful at first but can be harmful if they last too long. Long-term stress, caused by hormones like cortisol and adrenaline, weakens the immune system and makes people more likely to get sick. Important brain chemicals like serotonin and norepinephrine help control how our immune system works. Also, the connection between our gut and brain is an important way that mental health affects how our immune system functions. Getting older and experiencing stress early in life can affect how our immune system works. Inflammation caused by stress is connected to health issues like heart disease, depression, and autoimmune diseases. There are ways to manage stress, like being mindful and having support from friends, are important for keeping your immune system healthy and lessening harm caused by stress.
Collapse
Affiliation(s)
- Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| |
Collapse
|
20
|
Bosenberg M. Advances in Studying Cancer Immunology in Mice. Cold Spring Harb Perspect Med 2025; 15:a041682. [PMID: 38772704 PMCID: PMC11875087 DOI: 10.1101/cshperspect.a041682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
The recent rise in effective immuno-oncology therapies has increased demand for experimental approaches to model anticancer immunity. A variety of mouse models have been developed and used to study cancer immunology. These include mutagen-induced, genetically engineered, syngeneic, and other models of cancer immunology. These models each have the potential to define mechanistic aspects of anticancer immune responses, identify potential therapeutic targets, and serve as preclinical models for further therapeutic development. Specific benefits and liabilities are characteristic of particular cancer immunology modeling approaches. The optimal choice and utilization of models depends on the cancer immunology scientific question being addressed and can serve to increase mechanistic understanding and development of human immuno-oncology therapies.
Collapse
Affiliation(s)
- Marcus Bosenberg
- Departments of Dermatology, Pathology, and Immunobiology, Yale University, New Haven, Connecticut 06520, USA
| |
Collapse
|
21
|
Lee J, Jin Y, Wu W, Lee Y, Ha UH. Pseudomonas aeruginosa-derived DnaJ induces TLR2 expression through TLR10-mediated activation of the PI3K-SGK1 pathway in macrophages. Microbes Infect 2025:105481. [PMID: 39978578 DOI: 10.1016/j.micinf.2025.105481] [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: 11/08/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
TLR2 is a key component of the innate immune system, responsible for recognizing Gram-positive bacterial components and initiating inflammatory signaling cascades that activate defense responses. However, little is known about the regulatory effects of Pseudomonas aeruginosa (P. aeruginosa) on TLR2 expression. In this study, we investigated the potential link between P. aeruginosa-derived DnaJ and TLR2 expression in macrophages, as well as the activation of downstream signaling pathways. Our findings revealed that DnaJ significantly induced TLR2 expression in a dose- and time-dependent manner, predominantly affecting TLR2 with minimal impact on other TLRs, such as TLR4 and TLR5, which detect bacterial PAMPs. The DnaJ-mediated TLR2 induction was driven by activation of the PI3K-SGK1 signaling pathway, with TLR10 playing a crucial role in facilitating these effects. This increase in TLR2 expression led to enhanced production of inflammatory cytokines in response to secondary Staphylococcus aureus infections, indicating a role in boosting host defense mechanisms. In conclusion, these findings suggest that P. aeruginosa-derived DnaJ promotes TLR2 expression via TLR10-mediated activation of the PI3K-SGK1 pathway, thereby enhancing host immune responses against Gram-positive bacterial infections.
Collapse
Affiliation(s)
- Jaehoo Lee
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, 30019, Republic of Korea; Interdisciplinary Graduate Program for Artificial Intelligence Smart Convergence Technology, Korea University, Sejong, 30019, Republic of Korea
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, Nankai University, Tianjin, 300071, China
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, Nankai University, Tianjin, 300071, China
| | - Yeji Lee
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, 30019, Republic of Korea.
| | - Un-Hwan Ha
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, 30019, Republic of Korea; Interdisciplinary Graduate Program for Artificial Intelligence Smart Convergence Technology, Korea University, Sejong, 30019, Republic of Korea.
| |
Collapse
|
22
|
Tianyi FL, Ngari C, Wilkinson M, Parkurito S, Chebet E, Mumo E, Trelfa A, Otundo D, Crittenden E, Kephah GM, Harrison RA, Stienstra Y, Casewell NR, Lalloo DG, Oluoch GO. Clinical features of puff adder envenoming: case series of Bitis arietans snakebites in Kenya and a scoping review of the literature. PLoS Negl Trop Dis 2025; 19:e0012845. [PMID: 39928662 PMCID: PMC11828387 DOI: 10.1371/journal.pntd.0012845] [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: 08/18/2024] [Revised: 02/14/2025] [Accepted: 01/16/2025] [Indexed: 02/12/2025] Open
Abstract
INTRODUCTION The puff adder (Bitis arietans) is a medically important snake species found across much of Africa, yet there is limited literature on the clinical features and pathophysiology of envenoming after a puff adder bite. METHODS We conducted a case-series study to describe the clinical features of patients with puff adder bites who were treated in two primary healthcare facilities in Kenya and complemented our case-series with a scoping review of all published cases of puff adder envenoming that contained sufficient clinical details to highlight the major features. RESULTS Between December 2020 and September 2021, 15 patients were admitted with a suspected puff adder bite (based on the patient's description of the biting snake or confirmed in patients who brought the dead snake or a picture of the biting snake for identification) at the Chemolingot and Mwingi sub-county hospitals in Baringo and Kitui counties, central Kenya. Common local and systemic features on admission included pain (n=15, 100%), swelling (n=14, 93%), and haemorrhage (n=9, 60%). Coagulopathy (n=2, 13%), blistering (n=1, 8%) and shock (n=1, 8%) were less common. In addition, we conducted a literature review and identified 23 studies with detailed descriptions of the clinical features of puff adder envenoming from 37 patients. Local features were common and consistent across cases-swelling (100%, n=37) and pain (95%, n=35). Systemic features were less consistent, with 10 (27%) patients exhibiting hypotension on admission, 10 (27%) patients reporting a fever, and 13 (35%) developing anaemia. Some complications were more common in patients with bites by captive snakes (amputations), compared to patients with bites by wild snakes (hypotension). Snake identification was easier and more accurate after bites by captive snakes, but more challenging for patients bitten in community settings. CONCLUSION We combined clinical cases and a literature review to describe the common and less common clinical features of puff adder envenoming. Further clinical research incorporating serial laboratory assays of patients with definitively identified puff adder bites is crucial to better understand the pathophysiology of envenoming by this medically important snake species.
Collapse
Affiliation(s)
- Frank-Leonel Tianyi
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
| | - Cecilia Ngari
- Kenya Snakebite Research and Intervention Centre, Kenya Institute of Primate Research, Ministry of Health, Nairobi, Kenya,
| | - Mark Wilkinson
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
| | - Stanley Parkurito
- Kenya Snakebite Research and Intervention Centre, Kenya Institute of Primate Research, Ministry of Health, Nairobi, Kenya,
| | | | - Evans Mumo
- Chemolingot sub-county hospital, Baringo County, Kenya,
| | - Anna Trelfa
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
| | - Denis Otundo
- Kenya Snakebite Research and Intervention Centre, Kenya Institute of Primate Research, Ministry of Health, Nairobi, Kenya,
| | - Edouard Crittenden
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
| | - Geoffrey Maranga Kephah
- Kenya Snakebite Research and Intervention Centre, Kenya Institute of Primate Research, Ministry of Health, Nairobi, Kenya,
| | - Robert A. Harrison
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
| | - Ymkje Stienstra
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
- Kenya Snakebite Research and Intervention Centre, Kenya Institute of Primate Research, Ministry of Health, Nairobi, Kenya,
- Department of Internal Medicine/Infectious Diseases, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Nicholas R. Casewell
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
| | - David G. Lalloo
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
| | - George O. Oluoch
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke, United Kingdom,
- Kenya Snakebite Research and Intervention Centre, Kenya Institute of Primate Research, Ministry of Health, Nairobi, Kenya,
| |
Collapse
|
23
|
Li ET, Ji JY, Kong WJ, Shen DX, Li C, An CJ. A C-type lectin with dual carbohydrate recognition domains functions in innate immune response in Asian corn borer, Ostrinia furnacalis. INSECT SCIENCE 2025; 32:172-192. [PMID: 38772748 DOI: 10.1111/1744-7917.13382] [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: 11/05/2023] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/23/2024]
Abstract
C-type lectins (CTLs) act as pattern recognition receptors (PRRs) to initiate the innate immune response in insects. A CTL with dual carbohydrate recognition domains (CRDs) (named immulectin-4 [IML-4]) was selected from the Ostrinia furnacalis transcriptome dataset for functional studies. We cloned the full-length complementary DNA of O. furnacalis IML-4 (OfIML-4). It encodes a 328-residue protein with a Glu-Pro-Asn (EPN) and Gln-Pro-Asp (QPD) motifs in 2 CRDs, respectively. OfIML-4 messenger RNA levels increased significantly upon the bacterial and fungal infection. Recombinant OfIML-4 (rIML-4) and its individual CRDs (rCRD1 and rCRD2) exhibited the binding ability to various microorganisms including Escherichia coli, Micrococcus luteus, Pichia pastoris, and Beauveria bassiana, and the cell wall components including lipopolysaccharide from E. coli, peptidoglycan from M. luteus or Bacillus subtilis, and curdlan from Alcaligenes faecalis. The binding further induced the agglutination of E. coli, M. luteus, and B. bassiana in the presence of calcium, the phagocytosis of Staphylococcus aureus by the hemocytes, in vitro encapsulation and melanization of nickel-nitrilotriacetic acid beads, and a significant increase in phenoloxidase activity of plasma. In addition, rIML-4 significantly enhanced the phagocytosis, nodulation, and resistance of O. furnacalis to B. bassiana. Taken together, our results suggest that OfIML-4 potentially works as a PRR to recognize the invading microorganisms, and functions in the innate immune response in O. furnacalis.
Collapse
Affiliation(s)
- Er-Tao Li
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jia-Yue Ji
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, China
- Pomology Institute, Shanxi Agricultural University, Jinzhong, Shanxi Province, China
| | - Wei-Jie Kong
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, China
| | - Dong-Xu Shen
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, China
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
| | - Cai Li
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, China
| | - Chun-Ju An
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
24
|
Ahn MH, Choi YS, Lee SW, Choi S, Kim HA. Toll-Like Receptor Blockage by TIP-1 and MIP-2 Treatment Mitigates Inflammation in a Mouse Model of Adult-Onset Still's Disease or Still's Disease. Eur J Immunol 2025; 55:e202451227. [PMID: 39931754 DOI: 10.1002/eji.202451227] [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: 04/30/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 05/08/2025]
Abstract
The inflammatory response triggered by Toll-like receptors (TLRs) may implicated in the development of the pathogenesis of adult-onset Still's disease (AOSD). This study evaluated the efficacy of TLR inhibitor peptides, specifically TLR inhibitor peptide 1 (TIP-1) and MAL/MyD88 inhibitory peptide 2 (MIP-2) in animal models of AOSD. THP-1 cells were stimulated with TLR agonists and treated with TIP-1 or MIP-2. Interferon (IFN)-γ knock-out mice were induced with AOSD-like symptoms using Mycobacterium mixed with Freund's complete adjuvant (CFA), then treated with the peptides. THP-1 cells treated with TIP-1 and MIP-2 showed significantly decreased expression of TLRs agonist-induced MyD88 and phosphorylated NF-κB, except TLR9 agonists. Furthermore, the peptides resulted in a significant decrease in the concentrations of interleukin (IL)-1β and IL-6 in the culture supernatants, except TLR9 agonists. In animal models of AOSD, treatment with inhibitor peptides significantly improved their clinical symptoms. The administration of these peptides resulted in a significant decrease in serum levels of IL-1β and IL-18. The expression of inflammatory cytokines were downregulated in the spleen and lymph node of TIP-1 and MIP-2 treated mice. These findings suggest that TIP-1 and MIP-2 may be effective candidates for AOSD treatment, as they have broad specificity for TLRs.
Collapse
Affiliation(s)
- Mi-Hyun Ahn
- Department of Rheumatology, Ajou University School of Medicine, Suwon, South Korea
| | - Yang Seon Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Sang-Won Lee
- Department of Rheumatology, Ajou University School of Medicine, Suwon, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Hyoun-Ah Kim
- Department of Rheumatology, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
25
|
Bano Y, Shrivastava A, Shukla P, Chaudhary AA, Khan SUD, Khan S. The implication of microbiome in lungs cancer: mechanisms and strategies of cancer growth, diagnosis and therapy. Crit Rev Microbiol 2025; 51:128-152. [PMID: 38556797 DOI: 10.1080/1040841x.2024.2324864] [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: 07/17/2023] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 04/02/2024]
Abstract
Available evidence illustrates that microbiome is a promising target for the study of growth, diagnosis and therapy of various types of cancer. Lung cancer is a leading cause of cancer death worldwide. The relationship of microbiota and their products with diverse pathologic conditions has been getting large attention. The novel research suggests that the microbiome plays an important role in the growth and progression of lung cancer. The lung microbiome plays a crucial role in maintaining mucosal immunity and synchronizing the stability between tolerance and inflammation. Alteration in microbiome is identified as a critical player in the progression of lung cancer and negatively impacts the patient. Studies suggest that healthy microbiome is essential for effective therapy. Various clinical trials and research are focusing on enhancing the treatment efficacy by altering the microbiome. The regulation of microbiota will provide innovative and promising treatment strategies for the maintenance of host homeostasis and the prevention of lung cancer in lung cancer patients. In the current review article, we presented the latest progress about the involvement of microbiome in the growth and diagnosis of lung cancer. Furthermore, we also assessed the therapeutic status of the microbiome for the management and treatment of lung cancer.
Collapse
Affiliation(s)
- Yasmin Bano
- Department of Biotechnology, College of Life Sciences, Cancer Hospital and research Institute, Gwalior, India
- Centre for Genomics, Molecular and Human Genetics, Jiwaji University, Gwalior, India
| | - Abhinav Shrivastava
- Department of Biotechnology, College of Life Sciences, Cancer Hospital and research Institute, Gwalior, India
| | - Piyush Shukla
- Centre for Genomics, Molecular and Human Genetics, Jiwaji University, Gwalior, India
- Laboratory of Natural Products, Department of Rural Technology and Social Development, Guru Ghasidas University, Bilaspur, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Shahanavaj Khan
- Department of Medical Lab Technology, Indian Institute of Health Technology (IIHT), Deoband, Saharanpur, UP, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, Australia
| |
Collapse
|
26
|
da Silva Júnior WF, de Oliveira Costa KM, Castro Oliveira HM, Antunes MM, Mafra K, Nakagaki BN, Corradi da Silva PS, Megale JD, de Sales SC, Caixeta DC, Martins MM, Sabino-Silva R, de Paula CMP, Goulart LR, Rezende RM, Menezes GB. Physiological accumulation of lipid droplets in the newborn liver during breastfeeding is driven by TLR4 ligands. J Lipid Res 2025; 66:100744. [PMID: 39814317 PMCID: PMC11849619 DOI: 10.1016/j.jlr.2025.100744] [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: 07/17/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
The liver plays a central role in fat storage, but little is known about physiological fat accumulation during early development. Here we investigated a transient surge in hepatic lipid droplets observed in newborn mice immediately after birth. We developed a novel model to quantify liver fat content without tissue processing. Using high-resolution microscopy assessed the spatial distribution of lipid droplets within hepatocytes. Lugol's iodine staining determined the timing weaning period, and milk deprivation experiments investigated the relationship between milk intake and fat accumulation. Lipidomic analysis revealed changes in the metabolic profile of the developing liver. Finally, we investigated the role of Toll-like receptor 4 (TLR4) signaling in fat storage using knockout mice and cell-specific deletion strategies. Newborn mice displayed a dramatic accumulation of hepatic lipid droplets within the first 12 h after birth, persisting for the initial two weeks of life. This pattern coincided with exclusive milk feeding and completely abated by the third week, aligning with weaning. Importantly, the observed fat accumulation shared characteristics with established models of pathological steatosis, suggesting potential biological relevance. Lipid droplets were primarily localized within the cytoplasm of hepatocytes. Milk deprivation experiments demonstrated that milk intake is the primary driver of this transient fat accumulation. Lipidomic analysis revealed significant changes in the metabolic profile of newborn livers compared to adults. Interestingly, several highly abundant lipids in newborns were identified as putative ligands for TLR4. Subsequent studies using TLR4-deficient mice and cell-specific deletion revealed that TLR4 signaling, particularly within hepatocytes, plays a critical role in driving fat storage within the newborn liver. Additionally, a potential collaboration between metabolic and immune systems was suggested by the observed effects of myeloid cell-specific TLR4 ablation. This study demonstrates a unique phenomenon of transient hepatic fat accumulation in newborn mice driven by milk intake and potentially regulated by TLR4 signaling, particularly within hepatocytes.
Collapse
Affiliation(s)
- Wanderson Ferreira da Silva Júnior
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karen Marques de Oliveira Costa
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Hortência Maciel Castro Oliveira
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maísa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kassiana Mafra
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Brenda Naemi Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro Sérgio Corradi da Silva
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Júlia Duarte Megale
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sarah Campos de Sales
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Douglas Carvalho Caixeta
- Innovation Center in Salivary Diagnostics and Nanobiotechnology, Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | - Mário Machado Martins
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, Brazil
| | - Robinson Sabino-Silva
- Innovation Center in Salivary Diagnostics and Nanobiotechnology, Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | - Cristina Maria Pinto de Paula
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | - Rafael Machado Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
27
|
Li X, Wang X, Shang Z, Yang S, Tang Y, Xu W. Non-Immune Functions of Innate Immunity Acting on Physiological Processes: Insights from Drosophila. Int J Mol Sci 2025; 26:1087. [PMID: 39940855 PMCID: PMC11817114 DOI: 10.3390/ijms26031087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
As the first line of host immune defense, innate immunity plays a key role in warding off foreign pathogens and damage. Drosophila melanogaster, as a classical model animal for more than 100 years, is an important research model for studying innate immunity. In recent years, scientists have made remarkable progress in the recognition mechanisms of innate immunity, the mechanisms of effector molecules, and the modes of their response at the cellular and tissue levels. However, the interaction between innate immunity and other physiological functions remains relatively novel and has yet to be systematically explored. Here, we first briefly discuss the link between the innate immunity system and physiological regulation, from several representative perspectives such as sleep, insulin, and brain function. Then, using Drosophila as a model, we provide an overview of the physiological system and specifically summarize the research on the regulation of physiology by innate immunity, covering sleep, lipid metabolism, development, neurodegenerative diseases, memory, feeding, lifespan, movement, and antioxidation. This review provides valuable perspectives into how innate immunity influences other physiological processes, providing a deeper understanding of the complex roles underlying innate immunity.
Collapse
Affiliation(s)
| | | | | | | | | | - Wenhua Xu
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao 266071, China
| |
Collapse
|
28
|
Liu M, Fan G, Meng L, Yang K, Liu H. New perspectives on microbiome-dependent gut-brain pathways for the treatment of depression with gastrointestinal symptoms: from bench to bedside. J Zhejiang Univ Sci B 2025; 26:1-25. [PMID: 39428337 PMCID: PMC11735910 DOI: 10.1631/jzus.b2300343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 08/29/2023] [Indexed: 10/22/2024]
Abstract
Patients with depression are more likely to have chronic gastrointestinal (GI) symptoms than the general population, but such symptoms are considered only somatic symptoms of depression and lack special attention. There is a chronic lack of appropriate diagnosis and effective treatment for patients with depression accompanied by GI symptoms, and studying the association between depression and GI disorders (GIDs) is extremely important for clinical management. There is growing evidence that depression is closely related to the microbiota present in the GI tract, and the microbiota-gut-brain axis (MGBA) is creating a new perspective on the association between depression and GIDs. Identifying and treating GIDs would provide a key opportunity to prevent episodes of depression and may also improve the outcome of refractory depression. Current studies on depression and the microbially related gut-brain axis (GBA) lack a focus on GI function. In this review, we combine preclinical and clinical evidence to summarize the roles of the microbially regulated GBA in emotions and GI function, and summarize potential therapeutic strategies to provide a reference for the study of the pathomechanism and treatment of depression in combination with GI symptoms.
Collapse
Affiliation(s)
- Menglin Liu
- The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Brain Disease Regional Diagnosis and Treatment Center, Zhengzhou 450000, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301600, China
| | - Genhao Fan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301600, China
- The First Affiliated Hospital of Zhengzhou University, Department of Geriatrics, Zhengzhou 450052, China
| | - Lingkai Meng
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300131, China
| | - Kuo Yang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300131, China
| | - Huayi Liu
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300131, China.
| |
Collapse
|
29
|
Hanna MI, El Sayed AT, El Megeed OHA, Ibrahim MA, Korany RMS, Attia MM. Assessment of a new protocol strategy to control the ectoparasitic infestation in Nile tilapia (Oreochromis niloticus) using efficient natural products. BMC Vet Res 2025; 21:15. [PMID: 39799323 PMCID: PMC11724587 DOI: 10.1186/s12917-024-04387-z] [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: 04/27/2024] [Accepted: 11/18/2024] [Indexed: 01/15/2025] Open
Abstract
This study aimed to evaluate alternative in vivo treatment trials using natural products for ectoparasitic infestation on Nile tilapia; these two products were not previously used in the treatment of parasitic fish diseases. So, a total of 400 Oreochromis niloticus (O. niloticus) fish measured 10-15 cm in length; 350 from a fish farm in (Kafr Elsheikh and 50 from Nile River (Al Bahr Al Aazam), Egypt. The examined fishes were 10-15 ± 0.5 cm long and weighed from 45 g ± 5. The collected fish were examined for different clinical abnormalities. Each part of the fish underwent a careful microscopic examination of mucous surrounding the skin; gills and fins. Two feed supplements were used experimentally to decrease mortality and treat fish against ectoparasites (Herb-All PARA-X® and Herb-All CALM®). Total mRNA was extracted from the gills of different examined groups. Glucose; nitric oxide; cortisol as well as lysozyme activity were assessed in all groups. The gills of the examined fish were collected for histopathological examination. Only, Dactyolgyrus sp. was recovered. The intensity of the parasite was counted per microscopic field. The treated groups showed low levels of the parameters compared to the control positive group. Up-regulation of both Tumor necrosis factor-α (TNF- α) and heat shock protein 70 (hsp-70) were detected in fins, gills, and skin in the infested tilapia. The treatment and prophylaxis significantly downregulated both genes in the studied organ in a dose-dependent manner. Recorded lesions which were scored according to their severity. In conclusion; following the use of those products, fish health has been greatly improved and that is indicated by findings of immune reactions as well as histopathology.
Collapse
Affiliation(s)
- Magdy I Hanna
- Department of Aquatic Animal Medicine and Management, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Aya T El Sayed
- Department of Aquatic Animal Medicine and Management, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Ola Hasan Abd El Megeed
- Department of Aquatic Animal Medicine and Management, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Marwa A Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Reda M S Korany
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
- Department of Pathology, Faculty of Veterinary Medicine, Egyptian Chinese University, Cairo, Egypt
| | - Marwa M Attia
- Department of Parasitology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| |
Collapse
|
30
|
Lei S, Liu Y. Identifying the Involvement of Gut Microbiota in Retinal Vein Occlusion by Mendelian Randomization and Genetic Correlation Analysis. Transl Vis Sci Technol 2025; 14:5. [PMID: 39786739 PMCID: PMC11725986 DOI: 10.1167/tvst.14.1.5] [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: 08/24/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Purpose Previous researches have suggested an important association between gut microbiota (GM) and vascular pathologies such as atherosclerosis. This study aimed to explore the association between 196 GM taxa and retinal vein occlusion (RVO). Methods This study used Mendelian randomization (MR), linkage disequilibrium score regression (LDSC), and polygenic overlap analysis. Genome-wide association study (GWAS) data associated with 196 GM taxa was obtained from the MiBioGen consortium, involving a large number of European-ancestry participants. GWAS data of RVO was obtained from the FinnGen consortium and another study that also involved European-ancestry participants. Inverse-variance weighted was used as the primary approach for MR estimation. Moreover, LDSC and polygenic overlap analyses were performed to evaluate the genetic correlation between GM taxa and RVO. Results The MR results identified the association of six GM taxa, including class Bacilli, order Lactobacillales, family Streptococcaceae, genus Clostridium innocuum group, genus Family XIII AD3011 group, and genus Subdoligranulum with the development of RVO. In addition, the polygenic overlap analysis supported the genetic association between GM and RVO. Conclusions Our findings confirmed the association between six GM taxa and the development of RVO, thereby highlighting the effects of GM on retinal vascular health. Translational Relevance The results may provide the rationale for developing GM-based strategies for preventing the onset of RVO.
Collapse
Affiliation(s)
- Shizhen Lei
- Department of Ophthalmology, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Yani Liu
- Department of Otolaryngology & Head and Neck Surgery, Wuhan No.1 Hospital, Wuhan, Hubei, China
| |
Collapse
|
31
|
Abdulrahim AO, Doddapaneni NSP, Salman N, Giridharan A, Thomas J, Sharma K, Abboud E, Rochill K, Shreelakshmi B, Gupta V, Lakkimsetti M, Mowo-Wale A, Ali N. The gut-heart axis: a review of gut microbiota, dysbiosis, and cardiovascular disease development. Ann Med Surg (Lond) 2025; 87:177-191. [PMID: 40109640 PMCID: PMC11918638 DOI: 10.1097/ms9.0000000000002789] [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: 06/25/2024] [Accepted: 11/20/2024] [Indexed: 03/22/2025] Open
Abstract
Background Cardiovascular diseases (CVDs) are a major cause of morbidity and mortality worldwide and there are strong links existing between gut health and cardiovascular health. Gut microbial diversity determines gut health. Dysbiosis, described as altered gut microbiota, causes bacterial translocations and abnormal gut byproducts resulting in systemic inflammation. Objective To review the current literature on the relationships between gut microbiota, dysbiosis, and CVD development, and explore therapeutic methods to prevent dysbiosis and support cardiovascular health. Summary Dysbiosis increases levels of pro-inflammatory substances while reducing those of anti-inflammatory substances. This accumulative inflammatory effect negatively modulates the immune system and promotes vascular dysfunction and atherosclerosis. High Firmicutes to Bacteroidetes ratios, high trimethylamine-n-oxide to short-chain fatty acid ratios, high indole sulfate levels, low cardiac output, and polypharmacy are all associated with worse cardiovascular outcomes. Supplementation with prebiotics and probiotics potentially alleviates some CVD risk. Blood and stool samples may be used in clinical practice to quantify and qualify gut bacterial ratios and byproducts, assess patients' risk for adverse cardiovascular outcomes, and track their gut health progress. Further research is required to set population-based cutoffs for normal and abnormal gut microbiota and byproduct ratios.
Collapse
Affiliation(s)
| | | | - Nadhra Salman
- Department of Internal Medicine, Baqai Medical University, Karachi, Pakistan
| | | | | | - Kavya Sharma
- Maharishi Markandeshwar Medical College and Hospital, Himachal Pradesh, India
| | - Elias Abboud
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | | | - B Shreelakshmi
- Navodaya Medical College Hospital & Research Centre, Karnataka, India
| | | | | | | | - Noor Ali
- Dubai Medical College, Dubai, United Arab Emirates
| |
Collapse
|
32
|
Saiz-Ladera C. Generation of a Mouse Model for the Study of Thyroid Hormones Regulatory Effect on the Immune System. Methods Mol Biol 2025; 2876:61-75. [PMID: 39579308 DOI: 10.1007/978-1-0716-4252-8_4] [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: 11/25/2024]
Abstract
The generation of hypothyroid and hyperthyroid mouse models is one of the approaches used to investigate the complex interplay between thyroid hormones and the immune system. We present a detailed protocol describing how to induce endotoxic shock by lipopolysaccharide (LPS) administration, and how to investigate the role of immune populations, specifically macrophages, responding to endotoxemia.This book chapter provides the use of different molecular techniques, such as Western Blotting, Immunohistochemistry, q-PCR, Luciferase assays, or ChIP assays, with which researchers can gain valuable insights into the immune system's interaction with hormonal signaling pathways, for instance, examining the effect of thyroid hormones on signaling of STAT3, NF-κB, and ERK in response to LPS, and inflammatory mediators, such as interleukin-6 (IL-6) or tumor necrosis factor-alpha (TNFα) within these cells. The signaling pathways involved and the exploration of the relationship between thyroid hormones and the immune system can be analyzed using several molecular biology technologies in order to clarify their interplay in various disease states.
Collapse
Affiliation(s)
- Cristina Saiz-Ladera
- Unidad de Oncogenómica, Servicio de Oncohematología, Fundación para la Investigación Biomédica del Hospital Universitario Niño Jesús, Madrid, Spain.
| |
Collapse
|
33
|
Li C, Ai H, Zhang B, Huang X, Li B. C-type lectin 9 participates in the immune response, development and reproduction of Tribolium castaneum. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2025; 207:106223. [PMID: 39672654 DOI: 10.1016/j.pestbp.2024.106223] [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: 08/18/2024] [Revised: 11/18/2024] [Accepted: 11/24/2024] [Indexed: 12/15/2024]
Abstract
C-type lectins (CTLs), as a large family of pattern recognition receptors (PRRs), have been reported to be involved in bacterial infection, but the role of CTLs in development has been poorly understood in insects. The orthologues of Tribolium castaneum CTL9 (TcCTL9) have been identified among insects, but its functions were currently unclear. Therefore, we performed functional analysis of TcCTL9 in this study. Our results indicated that TcCTL9 could bind to bacteria through lipopolysaccharide and peptidoglycan, and agglutinate Gram-positive and Gram-positive bacteria in a Ca2+-dependent manner. Silencing TcCTL9 reduced the immune resistance to Staphylococcus aureus and Escherichia coli, decreased the expression of antimicrobial peptides and prophenoloxidase, and inhibited the phenoloxidase activity. These data suggested that TcCTL9 functioned in the immune response via the Toll and IMD pathways and prophenoloxidase system. During development, TcCTL9 had high expression in the periods of egg to larva and pupa to adult, and knockdown of TcCTL9 suppressed the metamorphosis, egg production and hatchability, and ovary development through ecdysone and juvenile hormone pathways in T. castaneum. This study comprehensively clarified the functions of TcCTL9 orthologues in insects and provided the theoretical basis for developing novel targets of pesticides.
Collapse
Affiliation(s)
- Chengjun Li
- College of Life Sciences, Nanjing Normal University, 210023 Nanjing, China
| | - Huayi Ai
- College of Life Sciences, Nanjing Normal University, 210023 Nanjing, China
| | - Biao Zhang
- College of Life Sciences, Nanjing Normal University, 210023 Nanjing, China
| | - Xiaoqiao Huang
- College of Life Sciences, Nanjing Normal University, 210023 Nanjing, China
| | - Bin Li
- College of Life Sciences, Nanjing Normal University, 210023 Nanjing, China.
| |
Collapse
|
34
|
Zhang X, Ling C, Xiong Z, Gong T, Luo S, Liu X, Zhang L, Liao C, Lu Y, Huang X, Zhou W, Zhou S, Liu Y, Tang J. Desuccinylation of TBK1 by SIRT5 regulates inflammatory response of macrophages in sepsis. Cell Rep 2024; 43:115060. [PMID: 39673708 DOI: 10.1016/j.celrep.2024.115060] [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: 12/21/2023] [Revised: 08/19/2024] [Accepted: 11/21/2024] [Indexed: 12/16/2024] Open
Abstract
Tank-binding kinase 1 (TBK1) is a critical signal transducer in the nuclear factor κB (NF-κB) and interferon regulatory factor (IRF) pathways, essential for innate immunity. However, its negative regulation mechanisms remain unclear. This study demonstrates that TBK1 succinylation, regulated by desuccinylase SIRT5, inhibits lipopolysaccharide (LPS)/Toll-like receptor 4 (TLR4)-mediated NF-κB and IRF signaling activation. We identified three key succinylation sites on TBK1: K38, K154, and K692. In endotoxemia and sepsis models, reduced SIRT5 levels in macrophages increased TBK1 succinylation, inhibiting its binding to IRF3 and TRAF2 and suppressing the inflammatory response. In vivo, adoptive transfer of macrophages expressing the succinylation-resistant TBK1-2KR (K154/692R) mutant reversed the inflammatory cytokine suppression caused by SIRT5 deficiency, exacerbating sepsis-induced lung injury. These findings reveal a novel mechanism by which SIRT5 modulates TBK1 activity and macrophage-mediated inflammation during sepsis.
Collapse
Affiliation(s)
- Xuedi Zhang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China; Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No. 1333, Xinhu Road, Baoan District, Shenzhen, Guangdong 518110, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chunxiu Ling
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Ziying Xiong
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Ting Gong
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No. 1333, Xinhu Road, Baoan District, Shenzhen, Guangdong 518110, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shuhua Luo
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Xiaolei Liu
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Lina Zhang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Chaoxiong Liao
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Yue Lu
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Xiao Huang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Wending Zhou
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China
| | - Shuangnan Zhou
- Senior Department of Infectious Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China.
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, No. 1333, Xinhu Road, Baoan District, Shenzhen, Guangdong 518110, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Jing Tang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; Guang Dong Medical University, Zhanjiang, Guangdong 524000, China.
| |
Collapse
|
35
|
Ramnani B, Devale T, Manivannan P, Haridas A, Malathi K. DHX15 and Rig-I Coordinate Apoptosis and Innate Immune Signaling by Antiviral RNase L. Viruses 2024; 16:1913. [PMID: 39772220 PMCID: PMC11680366 DOI: 10.3390/v16121913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
During virus infection, the activation of the antiviral endoribonuclease, ribonuclease L (RNase L), by a unique ligand 2'-5'-oilgoadenylate (2-5A) causes the cleavage of single-stranded viral and cellular RNA targets, restricting protein synthesis, activating stress response pathways, and promoting cell death to establish broad antiviral effects. The immunostimulatory dsRNA cleavage products of RNase L activity (RL RNAs) recruit diverse dsRNA sensors to activate signaling pathways to amplify interferon (IFN) production and activate inflammasome, but the sensors that promote cell death are not known. In this study, we found that DEAH-box polypeptide 15 (DHX15) and retinoic acid-inducible gene I (Rig-I) are essential for apoptosis induced by RL RNAs and require mitochondrial antiviral signaling (MAVS), c-Jun amino terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK) for caspase-3-mediated intrinsic apoptosis. In RNase L-activated cells, DHX15 interacts with Rig-I and MAVS, and cells lacking MAVS expression were resistant to apoptosis. RL RNAs induced the transcription of genes for IFN and proinflammatory cytokines by interferon regulatory factor 3 (IRF-3) and nuclear factor kB (NF-kB), while cells lacking both DHX15 and Rig-I showed a reduced induction of cytokines. However, apoptotic cell death is independent of both IRF-3 and NF-kB, suggesting that cytokine and cell death induction by RL RNAs are uncoupled. The RNA binding of both DHX15 and Rig-I is required for apoptosis induction, and the expression of both single proteins in cells lacking both DHX15 and Rig-I is insufficient to promote cell death by RL RNAs. Cell death induced by RL RNAs suppressed Coxsackievirus B3 (CVB3) replication, and inhibiting caspase-3 activity or cells lacking IRF-3 showed that the induction of apoptosis directly resulted in the CVB3 antiviral effect, and the effects were independent of the role of IRF-3.
Collapse
Affiliation(s)
- Barkha Ramnani
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Trupti Devale
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
| | - Praveen Manivannan
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Aiswarya Haridas
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
| | - Krishnamurthy Malathi
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
| |
Collapse
|
36
|
Zhang J, Zhang L, Liu D, Shi H, Zhang X, Chen J, Yang X, Zeng M, Zhang J, Feng T, Zhu X, Jing Z, Ji Z, Shi D, Feng L. Helicase protein DDX11 as a novel antiviral factor promoting RIG-I-MAVS-mediated signaling pathway. mBio 2024; 15:e0202824. [PMID: 39470258 PMCID: PMC11633105 DOI: 10.1128/mbio.02028-24] [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: 07/04/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Type Ι interferon (IFN) production mediated by retinoic acid-inducible gene 1 (RIG-I) and mitochondrial antiviral signaling protein (MAVS) is essential for antiviral innate immune responses. Here, we report the identification of a novel co-sensor for cytosolic nucleic acids: DEAD/H-box helicase 11 (DDX11), a member of the DExD/H (Asp-Glu-x-Asp/His)-box helicase family. Knockdown or knockout of DDX11 attenuated the ability of cells to increase IFN-β, IFN-stimulated gene 56, and C-X-C motif chemokine ligand 10 in response to SeV and poly (I:C) by blocking the activation of TANK-binding kinase 1 and IFN regulatory factor 3. Nucleic acid sensing by DDX11 was independent of the stimulator of IFN genes but was dependent on RIG-I and MAVS. DDX11 regulated RIG-I-MAVS-mediated IFN signaling by specifically interacting with nucleic acid, RIG-I, and MAVS to enhance RIG-I-double-strand RNA and RIG-I-MAVS binding affinity. Overall, our results identified a critical role for DDX11 in the innate immune response and provided molecular insights into the mechanisms by which DDX11 recognized cytosolic nucleic acid and interacted with RIG-Ι and MAVS for potent IFN signaling and antiviral immunity. IMPORTANCE Innate immunity is the first and most rapid host defense against virus infection. Recognition of viral RNA by the retinoic acid-inducible gene 1 (RIG-I)-like receptors (RLRs) initiates innate antiviral immune responses. How the binding of viral RNA to and activation of the RLRs are regulated remains enigmatic. In this study, we identified DEAD/H-box helicase 11 (DDX11) as a positive regulator of the RIG-I-mitochondrial antiviral signaling protein (MAVS)-mediated signaling pathways. Mechanistically, we demonstrated that DDX11 bound to viral RNA, interacted with RIG-I, and promoted their binding to viral RNA. DDX11 also promoted the interaction between RIG-I and MAVS and activation of RIG-I-MAVS signaling. Overall, our results elucidate the role of DDX11 in RIG-I-MAVS-dependent signaling pathways and may shed light on innate immune gene regulation.
Collapse
Affiliation(s)
- Jiyu Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Liaoyuan Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Dakai Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hongyan Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xin Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jianfei Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiaoman Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Miaomiao Zeng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jialin Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tingshuai Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiaoyuan Zhu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhaoyang Jing
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhaoyang Ji
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Da Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Li Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
37
|
Elmanfi S, Onyedibe KI, Aryal UK, Könönen E, Sintim HO, Gürsoy UK. Activation of cellular responses by cyclic dinucleotides and porphyromonas gingivalis lipopolysaccharide: a proteomic study on gingival fibroblasts. J Oral Microbiol 2024; 17:2431453. [PMID: 39669221 PMCID: PMC11632945 DOI: 10.1080/20002297.2024.2431453] [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: 04/01/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 12/14/2024] Open
Abstract
Background Bacterial cyclic dinucleotides (CDNs), cyclic di-guanosine monophosphate (c-di-GMP), and cyclic di-adenosine monophosphate (c-di-AMP) upregulate interferon signaling proteins of human gingival fibroblasts (HGFs). However, the simultaneous effect of bacterial CDNs and lipopolysaccharides (LPS) on the HGF proteome is unknown. Aim The aim was to apply an unbiased proteomics approach to evaluate how simultaneous exposure to CDNs and Porphyromonas gingivalis (Pg) LPS affect the global proteome of HGFs. Methods The proteomic responses of HGFs were examined under three different treatment conditions (c-di-AMP+Pg LPS, c-di-GMP+Pg LPS, and Pg LPS alone) by label-free quantitative mass spectrometry analysis. Results Simultaneous exposure to CDNs and Pg LPS significantly upregulated innate immunity-related and interferon signaling-related proteins, such as ubiquitin-like protein ISG15 (ISG15), deoxynucleoside triphosphate triphosphohydrolase (SAMHD1), interferon regulatory factor 9 (IRF-9), interferon-induced GTP-binding protein Mx (MX)1, and MX2. Interferon signaling pathway was the most significantly regulated canonical pathway in both CDN treatment groups. Conclusion Simultaneous exposure to CDNs and Pg LPS stimulates the periodontal immune response by activating the anti-microbial cellular responses of HGFs with some notable differences from individual exposures.
Collapse
Affiliation(s)
- Samira Elmanfi
- Department of Periodontology, Institute of Dentistry, University of Turku, Turku, Finland
| | - Kenneth I. Onyedibe
- Department of Chemistry, Purdue University, West Lafayette, USA
- Purdue Institute for Inflammation, Immunology and Infectious Disease and Purdue Institute for Drug Discovery, Purdue University, West Lafayette, USA
- Department of Biomedical Sciences, Mercer University School of Medicine, MaconGA, USA
| | - Uma K. Aryal
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, USA
| | - Eija Könönen
- Department of Periodontology, Institute of Dentistry, University of Turku, Turku, Finland
| | - Herman O. Sintim
- Department of Chemistry, Purdue University, West Lafayette, USA
- Purdue Institute for Inflammation, Immunology and Infectious Disease and Purdue Institute for Drug Discovery, Purdue University, West Lafayette, USA
| | - Ulvi Kahraman Gürsoy
- Department of Periodontology, Institute of Dentistry, University of Turku, Turku, Finland
| |
Collapse
|
38
|
Ábrahám Á, Gyulai G, Mihály J, Horváth A, Dobay O, Varga Z, Kiss É, Horváti K. Optimizing lipopeptide bioactivity: The impact of non-ionic surfactant dressing. J Pharm Anal 2024; 14:101020. [PMID: 39881961 PMCID: PMC11774939 DOI: 10.1016/j.jpha.2024.101020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 01/31/2025] Open
Abstract
The aim of the research is to increase the applicability of lipopeptides as drugs. To this end, non-ionic triblock copolymers, namely poloxamers, were applied. The physico-chemical properties of poloxamers vary depending on the length of the blocks. In our study, we experimented with different types and systematically investigated the variation of the critical micelle concentration (CMC) of poloxamers at 25 and 37 °C in different media. In addition, the cytotoxicity of the different poloxamer micelles on three different cell lines was evaluated, and based on the results, Plur104, Plur123, and Plur127 were selected. Fatty acid elongated derivatives of a short antibacterial peptide (pL1), a medium-sized anticancer peptide (pCM15), and a branched-chain vaccine antigen (pATIPC) were used as lipopeptide models, and their formulations with the selected poloxamers were investigated. The solubility and homogeneity of the lipopeptides were significantly increased, and dynamic light scattering (DLS) measurements showed the formation of small particles of around 20 nm, which were well reproducible and storable. Similar homogenous micelle formation was observed after freeze-drying and reconstitution with water. The pL1 lipopeptide, formulated with the selected poloxamers, exhibited enhanced antibacterial activity with significantly reduced haemolytic side effects. The pCM15 peptide, when incorporated into poloxamer micelles, showed significantly enhanced cytotoxicity against tumor cells. Additionally, the internalization rate of poloxamer-formulated pATIPC peptide by antigen-presenting model cells exceeded that of the unformulated peptide. Our results demonstrate the potential of poloxamers as promising tools for the formulation of lipopeptides and for the optimization of their selectivity.
Collapse
Affiliation(s)
- Ágnes Ábrahám
- MTA-HUN-REN TTK Lendület “Momentum” Peptide-Based Vaccines Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
- Laboratory of Interfaces and Nanostructures, Institute of Chemistry, Eötvös Loránd University, Budapest, H-1117, Hungary
| | - Gergő Gyulai
- MTA-HUN-REN TTK Lendület “Momentum” Peptide-Based Vaccines Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
- Laboratory of Interfaces and Nanostructures, Institute of Chemistry, Eötvös Loránd University, Budapest, H-1117, Hungary
| | - Judith Mihály
- HUN-REN TTK Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| | - Andrea Horváth
- Institute of Medical Microbiology, Semmelweis University, Budapest, H-1085, Hungary
| | - Orsolya Dobay
- Institute of Medical Microbiology, Semmelweis University, Budapest, H-1085, Hungary
| | - Zoltán Varga
- HUN-REN TTK Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
- Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, H-1111, Hungary
| | - Éva Kiss
- Laboratory of Interfaces and Nanostructures, Institute of Chemistry, Eötvös Loránd University, Budapest, H-1117, Hungary
| | - Kata Horváti
- MTA-HUN-REN TTK Lendület “Momentum” Peptide-Based Vaccines Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| |
Collapse
|
39
|
Okada Y, Kajiyama K, Ishiguro C, Nonaka T, Komaki T, Kuga W, Komiyama N, Iguchi T, Horiuchi N, Uyama Y. Risk of neutropenia in psoriasis patients prescribed anti-IL-23 antibody in comparison with anti-IL-17 antibody or adalimumab based on real-world data from the MID-NET ® in Japan. J DERMATOL TREAT 2024; 35:2373826. [PMID: 38964751 DOI: 10.1080/09546634.2024.2373826] [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: 04/01/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND To evaluate the risk of neutropenia during treatment with anti-IL-23 antibodies in patients with psoriasis. METHOD We conducted an observational study with cohort design using MID-NET® in Japan. We identified patients with psoriasis who were newly prescribed anti-IL-23 antibodies, anti-IL-17-antibodies, adalimumab, or apremilast between January 1, 2009, and March 31, 2021. We estimated the adjusted hazard ratio (aHR) of anti-IL-23 antibodies compared to that of anti-IL-17 antibodies, adalimumab, or apremilast, for the risk of grade 2 (neutrophil count < 1,500/μL) or grade 3 (neutrophil count < 1,000/μL) neutropenia. RESULTS Overall, 287 patients on anti-IL-23 antibodies, 189 patients on anti-IL-17 antibodies, 293 patients on adalimumab, and 540 patients on apremilast were included. Compared with anti-IL-17 antibodies, the aHR (95% confidence interval (CI)) of anti-IL-23 antibodies was 0.83 (0.27-2.51) for grade 2 and 0.40 (0.02-7.60) for grade 3 neutropenia; that when compared with adalimumab was 0.76 (0.28-2.06) for grade 2 but was not calculated for grade 3 as no cases were found; and that compared with apremilast was 3.88 (0.62-24.48) for grade 2 and 0.43 (0.02-11.63) for grade 3 neutropenia. CONCLUSION No clear increase in the risk of neutropenia with anti-IL-23 antibodies was observed.
Collapse
Affiliation(s)
- Yusuke Okada
- Office of Pharmacovigilance II, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
- Office of Pharmacovigilance I, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Kazuhiro Kajiyama
- Office of Pharmacovigilance II, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
- Office of Pharmacovigilance I, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
- Office of Regulatory Science Research, Center for Regulatory Science, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Chieko Ishiguro
- Office of Medical Informatics and Epidemiology, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Takahiro Nonaka
- Office of Medical Informatics and Epidemiology, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Tomomi Komaki
- Office of Pharmacovigilance II, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Wataru Kuga
- Office of Pharmacovigilance II, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Noriyuki Komiyama
- Office of Pharmacovigilance II, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Toyotaka Iguchi
- Office of Pharmacovigilance II, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Naoya Horiuchi
- Office of Pharmacovigilance I, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Yoshiaki Uyama
- Office of Medical Informatics and Epidemiology, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
- Center for Regulatory Science, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| |
Collapse
|
40
|
Ahmad F, Ahmad S, Srivastav AK, Upadhyay TK, Husain A, Khubaib M, Kang S, Park MN, Kim B, Sharma R. "β-glucan signalling stimulates NOX-2 dependent autophagy and LC-3 associated autophagy (LAP) pathway". Int J Biol Macromol 2024; 282:136520. [PMID: 39401634 DOI: 10.1016/j.ijbiomac.2024.136520] [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: 08/02/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 12/19/2024]
Abstract
β-Glucan, a complex polysaccharide derived from fungal and yeast cell walls, plays a crucial role in modulating immune responses through their interaction with receptors such as Dectin-1 and Complement receptor 3 (CR-3). This review provides an in-depth analysis of the molecular mechanisms by which β-glucans activate receptor-mediated signalling pathways, focusing particularly on the LC3-associated phagocytosis (LAP) and autophagy pathways. Hence, we explore how β-glucan receptor engagement stimulates NADPH oxidase 2 (NOX-2), leading to the intracellular production of significant level of reactive oxygen species (ROS) essential for both conventional autophagy and LAP. While significant progress has been made in elucidation of downstream signaling by glucans, the regulation of phago-lysosomal maturation and antigen presentation during LAP induction still remains less explored. This review aims to provide a comprehensive overview of these pathways and their regulation by β-glucans. By consolidating the current knowledge, we seek to highlight how these mechanisms can be leveraged for therapeutic applications, particularly in the context of tuberculosis (TB) management, where β-glucans could serve as host-directed adjuvant therapies to combat drug-resistant strains. Despite major advancements in this field, currently key research gaps still persist, including detailed molecular interactions between β-glucan receptors and NOX-2 and the translation of these findings to in-vivo models and clinical investigations. This review underscores the need for further research to explore the therapeutic potential of β-glucans in managing not only tuberculosis but also other diseases such as cancer, cardiovascular conditions, and metabolic disorders.
Collapse
Affiliation(s)
- Firoz Ahmad
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India; Department of Physiological Sciences, Oklahoma Centre for Respiratory and Infectious Diseases, Oklahoma State University, OK 74074, United States of America
| | - Shad Ahmad
- Department of Biochemistry, Dr. Ram Manohar Lohia Avadh University, Faizabad 224001, Uttar Pradesh, India
| | - Anurag Kumar Srivastav
- Department of Clinical Immunology & Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| | - Tarun Kumar Upadhyay
- Department of Life Sciences, Parul Institute of Applied Sciences & Research and Development Cell, Parul University, Vadodara 391760, Gujarat, India
| | - Adil Husain
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India; Department of Biochemistry, Babu Banarasi Das [BBD] College of Dental Sciences BBD University, Lucknow 226028, Uttar Pradesh, India
| | - Mohd Khubaib
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea.
| | - Rolee Sharma
- Department of Life Sciences & Biotechnology, CSJM University, Kanpur 228024, Uttar Pradesh, India.
| |
Collapse
|
41
|
Hopo MG, Mabrok M, Abu-Elala N, Yu Y. Navigating Fish Immunity: Focus on Mucosal Immunity and the Evolving Landscape of Mucosal Vaccines. BIOLOGY 2024; 13:980. [PMID: 39765647 PMCID: PMC11727089 DOI: 10.3390/biology13120980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 01/15/2025]
Abstract
The growing role of aquaculture in global food security has underscored the need for advanced immunological insights to protect fish health and boost productivity. As aquaculture's importance rises, understanding fish immunity is crucial for developing effective vaccination strategies. Fish possess a specialized immune system with unique mucosal structures that enable resilience in aquatic environments. This review examines critical advances in fish mucosal immunity, particularly focusing on mucosal vaccines that target infection at primary entry points, such as the gills, skin, and gastrointestinal tract. Mucosal vaccination has demonstrated a compelling capacity to stimulate localized and systemic immune responses, offering enhanced protection against waterborne pathogens. Additionally, this review addresses knowledge gaps from previous research on the global aquaculture vaccines market by offering a regional perspective on industry developments, recent trends, and innovative vaccine formulations. In doing so, it highlights the role of mucosal vaccines in overcoming the specific challenges of fish farming and supporting sustainable aquaculture. This synthesis of current methodologies, industry practices, and future directions contributes to a deeper understanding of fish immunology, ultimately informing strategies to achieve optimal disease management and bolster global aquaculture resilience.
Collapse
Affiliation(s)
- Mai G. Hopo
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China;
| | - Mahmoud Mabrok
- Department of Fish Diseases and Management, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Department of Microbiology and Parasitology, Faculty of Veterinary Medicine, King Salman International University, Ras Sudr 46612, Egypt
| | - Nermeen Abu-Elala
- Department of Aquatic Animal Medicine and Management, Faculty of Veterinary Medicine, Cairo University, Cairo 12211, Egypt
- Department of Animal Medicine, Faculty of Veterinary Medicine, King Salman International University, Ras Sudr 46612, Egypt
| | - Yongyao Yu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China;
- Hubei Hongshan Laboratory, Wuhan 430070, China
| |
Collapse
|
42
|
Liu Y, Zhang J, Zhao H, Zhong F, Li J, Zhao L. VBNC Cronobacter sakazakii survives in macrophages by resisting oxidative stress and evading recognition by macrophages. BMC Microbiol 2024; 24:458. [PMID: 39506633 PMCID: PMC11539806 DOI: 10.1186/s12866-024-03595-9] [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: 05/03/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Survival in host macrophages is an effective strategy for pathogenic bacterial transmission and pathogenesis. Our previous study found that viable but non-culturable (VBNC) Cronobacter Sakazakii (C. sakazakii) can survive in macrophages, but its survival mechanism is not clear. In this study, we investigated the possible mechanisms of VBNC C. sakazakii survival in macrophages in terms of environmental tolerance within macrophages and evasion of macrophages recognition. The results revealed that VBNC C. sakazakii survived under oxidative conditions at a higher rate than the culturable C. sakazakii. Moreover, the stringent response gene (relA and spoT) and the antioxidant-related genes (sodA, katG, and trxA) were up-regulated, indicating that VBNC C. sakazakii may regulate antioxidation through stringent response. On the other hand, compared with culturable C. sakazakii, VBNC C. sakazakii caused reduced response (Toll-like receptor 4) in macrophages, which was attributed to the suppression of biosynthesis of the lipopolysaccharides (LPS). Furthermore, we found that ellagic acid can reduce the survival rate of bacteria in macrophages by improving the immune TLR4 recognition ability of macrophages. In conclusion, VBNC C. sakazakii may survive in macrophages by regulating oxidative tolerance through stringent response and altering LPS synthesis to evade TLR4 recognition by macrophages, which suggests the pathogenic risk of VBNC C. sakazakii.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Jingfeng Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Haoqing Zhao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Feifeng Zhong
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Jianyu Li
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Lichao Zhao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China.
| |
Collapse
|
43
|
Bhat MF, Srdanović S, Sundberg LR, Einarsdóttir HK, Marjomäki V, Dekker FJ. Impact of HDAC inhibitors on macrophage polarization to enhance innate immunity against infections. Drug Discov Today 2024; 29:104193. [PMID: 39332483 DOI: 10.1016/j.drudis.2024.104193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/26/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Innate immunity plays an important role in host defense against pathogenic infections. It involves macrophage polarization into either the pro-inflammatory M1 or the anti-inflammatory M2 phenotype, influencing immune stimulation or suppression, respectively. Epigenetic changes during immune reactions contribute to long-term innate immunity imprinting on macrophage polarization. It is becoming increasingly evident that epigenetic modulators, such as histone deacetylase (HDAC) inhibitors (HDACi), enable the enhancement of innate immunity by tailoring macrophage polarization in response to immune stressors. In this review, we summarize current literature on the impact of HDACi and other epigenetic modulators on the functioning of macrophages during diseases that have a strong immune component, such as infections. Depending on the disease context and the chosen therapeutic intervention, HDAC1, HDAC2, HDAC3, HDAC6, or HDAC8 are particularly important in influencing macrophage polarization towards either M1 or M2 phenotypes. We anticipate that therapeutic strategies based on HDAC epigenetic mechanisms will provide a unique approach to boost immunity against disease challenges, including resistant infections.
Collapse
Affiliation(s)
- Mohammad Faizan Bhat
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Sonja Srdanović
- Akthelia Pharmaceuticals, Grandagardi 16, 101 Reykjavik, Iceland
| | - Lotta-Riina Sundberg
- Department of Biological and Environmental Sciences and Nanoscience Center, 40014 University of Jyväskylä, Jyväskylä, Finland
| | | | - Varpu Marjomäki
- Department of Biological and Environmental Sciences and Nanoscience Center, 40014 University of Jyväskylä, Jyväskylä, Finland
| | - Frank J Dekker
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
44
|
Aboulaghras S, Bouyahya A, El Kadri K, Khalid A, Abdalla AN, Hassani R, Lee LH, Bakrim S. Protective and stochastic correlation between infectious diseases and autoimmune disorders. Microb Pathog 2024; 196:106919. [PMID: 39245422 DOI: 10.1016/j.micpath.2024.106919] [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: 08/10/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
A priori, early exposure to a wide range of bacteria, viruses, and parasites appears to fortify and regulate the immune system, potentially reducing the risk of autoimmune diseases. However, improving hygiene conditions in numerous societies has led to a reduction in these microbial exposures, which, according to certain theories, could contribute to an increase in autoimmune diseases. Indeed, molecular mimicry is a key factor triggering immune system reactions; while it seeks pathogens, it can bind to self-molecules, leading to autoimmune diseases associated with microbial infections. On the other hand, a hygiene-based approach aimed at reducing the load of infectious agents through better personal hygiene can be beneficial for such pathologies. This review sheds light on how the evolution of the innate immune system, following the evolution of molecular patterns associated with microbes, contributes to our protection but may also trigger autoimmune diseases linked to microbes. Furthermore, it addresses how hygiene conditions shield us against autoimmune diseases related to microbes but may lead to autoimmune pathologies not associated with microbes.
Collapse
Affiliation(s)
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco.
| | - Kawtar El Kadri
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco.
| | - Asaad Khalid
- Health Research Centre, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia.
| | - Rym Hassani
- Environment and Nature Research Centre, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; Biology Department, University College AlDarb, Jazan University, Jazan 45142, Saudi Arabia.
| | - Learn-Han Lee
- Microbiome Research Group, Research Centre for Life Science and Healthcare, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute (CBI), University of Nottingham Ningbo China, 315000, Ningbo, China; Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir, 80000, Morocco.
| |
Collapse
|
45
|
Yan YR, Sun YH. Genotypic diversity and immunological implications of porcine circovirus: Inspiration from PCV1 to PCV4. Microb Pathog 2024; 196:106997. [PMID: 39369754 DOI: 10.1016/j.micpath.2024.106997] [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: 04/10/2024] [Revised: 09/16/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Porcine circovirus (PCV) is a group of DNA viruses that cause diseases in pigs, with multiple genotypes ranging from PCV1 to PCV4. PCV1 is generally considered non-pathogenic, while PCV2 can cause severe immune system damage, especially associated with porcine multisystemic wasting syndrome (PMWS). PCV2 has a genetic homology of about 68 % but differs from PCV1 in antigenicity and phenotype. PCV3 and PCV4 have lower genetic homology with PCV1 and PCV2, with limited research available on their pathogenicity. During virus infection, the host's innate immune system detects PCVs through pattern recognition receptors (PRRs) like TLRs and NLRs. PCV disrupts immune pathways, including interferon and NF-κB pathways, aiding viral replication and causing immunosuppression. This review systematically compares the characteristics and pathogenicity of different genotypes of PCV and their interactions with the host's immune system, aiming to better understand the mechanisms of PCV infection and provide a theoretical basis for prevention and treatment.
Collapse
Affiliation(s)
- You-Rong Yan
- Jiangsu Agri-animal Husbandry Vocational College, No. 8 Fenghuang East Road, Hailing District, Taizhou City, Jiangsu Province, 225300, China.
| | - Ying-Hui Sun
- Shanghai Academy of Agricultural Sciences, No.2901 Beidi Road, Minhang District, Shanghai, 201106, China
| |
Collapse
|
46
|
Chen Z, Chen X, Zou Y, Zhou Y, Du J, Qin Y, Zou P, Zhang J, Zhu Y, Zhang Z, Wang Y. The immune function of TLR4-1 gene in Octopus sinensis revealed by RNAi and RNA-seq. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109899. [PMID: 39265964 DOI: 10.1016/j.fsi.2024.109899] [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: 06/11/2024] [Revised: 08/29/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Toll-like receptors (TLRs) are a class of conserved pattern recognition receptors (PRRs) that are crucial for initiating the innate immune response and aiding in the clearance of pathogenic organisms. Many studies have identified TLR4 as a distinctive member of the TLR family, capable of activating both the Myeloid differentiation factor 88-dependent signaling pathway (MyD88-dependent) and the TIR-domain-containing adaptor inducing IFN-β dependent signaling pathway (TRIF-dependent). Nevertheless, the role of TLR4 in Cephalopoda is still largely unexplored. To elucidate the immune function of the OsTLR4-1 gene in Octopus sinensis, the OsTLR4-1 gene was first validated and analyzed in this study. The cDNA comprises a 2475 bp ORF region, encoding 824 amino acids. Evolutionary tree analysis indicated a high homology and a close phylogenetic relationship between the Octopus sinensis and other mollusks. RNA interference (RNAi) experiments demonstrated that the expression level of OsTLR4-1 gene and its protein in the lymphocytes of the RNAi group treated with OsTLR4-1 dsRNA was extremely significantly lower than that of the blank control group and negative control group (P < 0.01), and the expression of downstream genes of OsTLR4-1, including ligand MyD88, IRAK4, TRAF6, MKK6, Hsp90, COX2, TRAF3, and RIP1, were significantly down-regulated compared to the blank and negative control group (P < 0.01). Additionally, OsTLR4-1 expression in lymphocytes was highly significantly up-regulated in the LPS-treated group compared to the blank control group (P < 0.01), while its expression was extremely significantly lower in the LPS-treated group after OsTLR4-1 interference than in the blank control group (P < 0.01). The expression of its downstream effector genes Big Defensin (Big-Def) and histone H2A.V (H2A.V) was highly significantly up-regulated in lymphocytes in the LPS-treated group compared to the blank control group (P < 0.01), while their expression in the LPS-treated group after OsTLR4-1 interference was extremely significantly lower than that in the blank control group (P < 0.01). Through comparative transcriptome analysis of the RNAi group and the blank control group, it was found that differentially expressed genes were enriched in the Toll-like receptor signaling pathway, PI3K-AKT signaling pathway, P53 signaling pathway, MAPK signaling pathway, and NF-κB signaling pathway. qRT-PCR results of key genes in these pathways revealed a decrease in all genes except IκB and Jun2 genes. This study enhances our understanding of the immune function of the TLR gene family in O. sinensis and provides a foundation for further research into innate immune signaling pathways in cephalopods.
Collapse
Affiliation(s)
- Zebin Chen
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Xinxin Chen
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Yihua Zou
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Yuquan Zhou
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Jiahui Du
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Yongjie Qin
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Pengfei Zou
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Jianming Zhang
- Putian Municipal Institute of Fishery Science, Putian, 351100, China
| | - Youfang Zhu
- Putian Municipal Institute of Fishery Science, Putian, 351100, China
| | - Ziping Zhang
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| | - Yilei Wang
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China.
| |
Collapse
|
47
|
Chen Y, Yang H, Wu X, Liu Z, Chen Y, Wei Q, Lin J, Yu Y, Tu Q, Li H. Interferon Regulatory Factors ( IRF1, IRF4, IRF5, IRF7 and IRF9) in Sichuan taimen ( Hucho bleekeri): Identification and Functional Characterization. Genes (Basel) 2024; 15:1418. [PMID: 39596618 PMCID: PMC11593489 DOI: 10.3390/genes15111418] [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: 09/28/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Interferon regulatory factors (IRFs) are multifunctional transcription factors that play important roles in the transcriptional regulation of interferons and in the immune response to pathogens. Therefore, studying the interferon system in fish is highly relevant in the prevention and treatment of viral diseases. Methods: In this study, five IRF genes (IRF1, IRF4, IRF5, IRF7 and IRF9) were identified and characterized in Hucho bleekeri, and their expression profiles were determined after LPS and Poly(I:C) treatment. Results: These IRFs have typical DNA-binding domains and IRF-association domains. Amino acid sequence comparison revealed high homology between these IRFs and those of other vertebrates, with the highest homology being with other salmonid fish. Phylogenetic analysis revealed that these IRFs are divided into four subfamilies (IRF1, IRF3, IRF4 and IRF5), with both IRF4 and IRF9 belonging to the IRF4 subfamily. IRF genes were widely expressed in all of the tested tissues, with IRF1, IRF4 and IRF9 being highly expressed in the spleen and kidney and IRF5 and IRF7 highly expressed in the gonads. IRF1, IRF4 and IRF5 expression was induced at different time points post-LPS challenge. IRF7 and IRF9 expression in the spleen and head kidney was not significantly altered by LPS induction. Poly(I:C) treatment altered IRF expression more significantly than LPS treatment. Poly(I:C) significantly altered the spleen and head kidney expression of all five IRFs. Conclusions: These findings reveal the potential role of IRFs in the antiviral response of H. bleekeri and provide a reference for examining signal transduction pathways in the interferon system in fish.
Collapse
Affiliation(s)
- Yeyu Chen
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Fish Resources and Environment, The Upper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu 611730, China
| | - Huanchao Yang
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Fish Resources and Environment, The Upper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu 611730, China
| | - Xiaoyun Wu
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Fish Resources and Environment, The Upper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu 611730, China
| | - Zhao Liu
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Fish Resources and Environment, The Upper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu 611730, China
| | - Yanling Chen
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Fish Resources and Environment, The Upper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu 611730, China
| | - Qinyao Wei
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Jue Lin
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Fish Resources and Environment, The Upper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu 611730, China
| | - Yi Yu
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Fish Resources and Environment, The Upper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu 611730, China
| | - Quanyu Tu
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Fish Resources and Environment, The Upper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu 611730, China
| | - Hua Li
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611730, China; (Y.C.); (H.Y.); (X.W.); (Z.L.); (Y.C.); (Q.W.); (J.L.); (Y.Y.); (Q.T.)
- Fish Resources and Environment, The Upper Reaches of the Yangtze River Observation and Research Station of Sichuan Province, Chengdu 611730, China
| |
Collapse
|
48
|
Vitaliti A, Reggio A, Palma A. Macrophages and autophagy: partners in crime. FEBS J 2024. [PMID: 39439196 DOI: 10.1111/febs.17305] [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: 09/02/2024] [Revised: 09/25/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Macrophages and autophagy are intricately linked, both playing vital roles in maintaining homeostasis and responding to disease. Macrophages, known for their 'eating' function, rely on a sophisticated digestion system to process a variety of targets, from apoptotic cells to pathogens. The connection between macrophages and autophagy is established early in their development, influencing both differentiation and mature functions. Autophagy regulates essential immune functions, such as inflammation control, pathogen clearance, and antigen presentation, linking innate and adaptive immunity. Moreover, it modulates cytokine production, ensuring a balanced inflammatory response that prevents excessive tissue damage. Autophagy also plays a critical role in macrophage polarization, influencing their shift between pro-inflammatory and anti-inflammatory states. This review explores the role of autophagy in macrophages, emphasizing its impact across various tissues and pathological conditions, and detailing the cellular and molecular mechanisms by which autophagy shapes macrophage function.
Collapse
Affiliation(s)
- Alessandra Vitaliti
- Department of Chemical Science and Technologies, "Tor Vergata" University of Rome, Italy
| | - Alessio Reggio
- Saint Camillus International University of Health Sciences, Rome, Italy
| | - Alessandro Palma
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Italy
| |
Collapse
|
49
|
Li W, Zhou J, Gu Y, Chen Y, Huang Y, Yang J, Zhu X, Zhao K, Yan Q, Zhao Z, Li X, Chen G, Jia X, Gao SJ, Lu C. Lactylation of RNA m 6A demethylase ALKBH5 promotes innate immune response to DNA herpesviruses and mpox virus. Proc Natl Acad Sci U S A 2024; 121:e2409132121. [PMID: 39413129 PMCID: PMC11513906 DOI: 10.1073/pnas.2409132121] [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: 05/08/2024] [Accepted: 08/31/2024] [Indexed: 10/18/2024] Open
Abstract
RNA N6-methyladenosine (m6A) demethylase AlkB homolog 5 (ALKBH5) plays a crucial role in regulating innate immunity. Lysine acylation, a widespread protein modification, influences protein function, but its impact on ALKBH5 during viral infections has not been well characterized. This study investigates the presence and regulatory mechanisms of a previously unidentified lysine acylation in ALKBH5 and its role in mediating m6A modifications to activate antiviral innate immune responses. We demonstrate that ALKBH5 undergoes lactylation, which is essential for an effective innate immune response against DNA herpesviruses, including herpes simplex virus type 1 (HSV-1), Kaposi's sarcoma-associated herpesvirus (KSHV), and mpox virus (MPXV). This lactylation attenuates viral replication. Mechanistically, viral infections enhance ALKBH5 lactylation by increasing its interaction with acetyltransferase ESCO2 and decreasing its interaction with deacetyltransferase SIRT6. Lactylated ALKBH5 binds interferon-beta (IFN-β) messenger RNA (mRNA), leading to demethylation of its m6A modifications and promoting IFN-β mRNA biogenesis. Overexpression of ESCO2 or depletion of SIRT6 further enhances ALKBH5 lactylation to strengthen IFN-β mRNA biogenesis. Our results identify a posttranslational modification of ALKBH5 and its role in regulating antiviral innate immune responses through m6A modification. The finding provides an understanding of innate immunity and offers a potential therapeutic target for HSV-1, KSHV, and MPXV infections.
Collapse
Affiliation(s)
- Wan Li
- Department of Gynecology, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing210004, People’s Republic of China
- Department of Microbiology, Nanjing Medical University, Nanjing211166, People’s Republic of China
- Changzhou Medical Center, Nanjing Medical University, Nanjing211166, People’s Republic of China
| | - Jing Zhou
- Department of Microbiology, Nanjing Medical University, Nanjing211166, People’s Republic of China
| | - Yang Gu
- Department of Microbiology, Nanjing Medical University, Nanjing211166, People’s Republic of China
| | - Yuheng Chen
- Department of Microbiology, Nanjing Medical University, Nanjing211166, People’s Republic of China
| | - Yiming Huang
- Department of Microbiology, Nanjing Medical University, Nanjing211166, People’s Republic of China
| | - Jingxin Yang
- Department of Microbiology, Nanjing Medical University, Nanjing211166, People’s Republic of China
| | - Xiaojuan Zhu
- Jiangsu Provincial Medical Key Laboratory of Pathogenic Microbiology in Emerging Major Infectious Diseases, National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing210009, People’s Republic of China
| | - Kangchen Zhao
- Jiangsu Provincial Medical Key Laboratory of Pathogenic Microbiology in Emerging Major Infectious Diseases, National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing210009, People’s Republic of China
| | - Qin Yan
- Department of Microbiology, Nanjing Medical University, Nanjing211166, People’s Republic of China
- Changzhou Medical Center, Nanjing Medical University, Nanjing211166, People’s Republic of China
| | - Zongzheng Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun130122, People’s Republic of China
| | - Xiao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun130122, People’s Republic of China
| | - Guochun Chen
- Changzhou Medical Center, Nanjing Medical University, Nanjing211166, People’s Republic of China
- Department of Infectious Diseases, Changzhou Third People’s Hospital, Changzhou213000, People’s Republic of China
| | - Xuemei Jia
- Department of Gynecology, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing210004, People’s Republic of China
| | - Shou-Jiang Gao
- Tumor Virology Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA15232
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA15232
| | - Chun Lu
- Department of Gynecology, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing210004, People’s Republic of China
- Department of Microbiology, Nanjing Medical University, Nanjing211166, People’s Republic of China
- Changzhou Medical Center, Nanjing Medical University, Nanjing211166, People’s Republic of China
| |
Collapse
|
50
|
Gómez-Oro C, Latorre MC, Arribas-Poza P, Ibáñez-Escribano A, Baca-Cornejo KR, Gallego-Valle J, López-Escobar N, Mondéjar-Palencia M, Pion M, López-Fernández LA, Mercader E, Pérez-Milán F, Relloso M. Progesterone promotes CXCl2-dependent vaginal neutrophil killing by activating cervical resident macrophage-neutrophil crosstalk. JCI Insight 2024; 9:e177899. [PMID: 39298265 PMCID: PMC11529979 DOI: 10.1172/jci.insight.177899] [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: 12/01/2023] [Accepted: 09/05/2024] [Indexed: 09/21/2024] Open
Abstract
Vaginal infections in women of reproductive age represent a clinical dilemma with significant socioeconomic implications. The current understanding of mucosal immunity failure during early pathogenic invasions that allows the pathogen to grow and thrive is far from complete. Neutrophils infiltrate most tissues following circadian patterns as part of normal repair, regulation of microbiota, or immune surveillance and become more numerous after infection. Neutrophils are responsible for maintaining vaginal immunity. Specific to the vagina, neutrophils continuously infiltrate at high levels, although during ovulation, they retreat to avoid sperm damage and permit reproduction. Here we show that, after ovulation, progesterone promotes resident vaginal macrophage-neutrophil crosstalk by upregulating Yolk sac early fetal organs (FOLR2+) macrophage CXCl2 expression, in a TNFA-patrolling monocyte-derived macrophage-mediated (CX3CR1hiMHCIIhi-mediated) manner, to activate the neutrophils' capacity to eliminate sex-transmitted and opportunistic microorganisms. Indeed, progesterone plays an essential role in conciliating the balance between the commensal microbiota, sperm, and the threat of pathogens because progesterone not only promotes a flurry of neutrophils but also increases neutrophilic fury to restore immunity after ovulation to thwart pathogenic invasion after intercourse. Therefore, modest progesterone dysregulations could lead to a suboptimal neutrophilic response, resulting in insufficient mucosal defense and recurrent unresolved infections.
Collapse
Affiliation(s)
- Carla Gómez-Oro
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Maria C. Latorre
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Patricia Arribas-Poza
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Alexandra Ibáñez-Escribano
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Katia R. Baca-Cornejo
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | | | - Natalia López-Escobar
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Mabel Mondéjar-Palencia
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marjorie Pion
- Laboratorio de InmunoRegulación, IiSGM, Madrid, Spain
| | - Luis A. López-Fernández
- Laboratorio de Farmacogenética, Grupo de Farmacia Hospitalaria y Farmacogenómica, IiSGM, Madrid, Spain
| | - Enrique Mercader
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Unidad Cirugía Endocrino-metabólica, Servicio de Cirugía General y Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Federico Pérez-Milán
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Unidad de Reproducción Asistida, Servicio de Obstetricia y Ginecología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Miguel Relloso
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|